Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 94
Filter
1.
Immunol Lett ; 239: 77-87, 2021 11.
Article in English | MEDLINE | ID: mdl-34508790

ABSTRACT

Cancer immunotherapy is emerging as a viable treatment option for several types of cancer. Active immunotherapy aims for the induction of specific antitumor immune responses; this goal requires strategies capable of increasing the immunogenicity of tumour antigens. Parvovirus B19 virus-like particles (B19-VLPs) formed of VP2 protein had been shown to be an effective multi-neoepitope delivery system capable of inducing specific cellular responses towards coupled antigens and reducing tumour growth and lung metastases in triple negative breast cancer mouse model. These findings encouraged us to further characterise these VP2 B19-VLPs by testing their capacity to simultaneously induce cellular and humoral responses towards other tumour-associated antigens, as this had not yet been evaluated. Here, we designed and evaluated in the 4T1 breast cancer model the prophylactic and therapeutic effect of VP2 B19-VLPs decorated with cellular (P53) and humoral (MUC1) epitopes. Balb/c mice were immunised with chimaeric VLPs, vehicle, or VLPs plus adjuvant. Tumour establishment and growth, lung metastasis, and cellular and humoral immune responses were evaluated. The prophylactic administration of chimaeric VLPs without adjuvant prevented the establishment of the tumour, while by therapeutic administration, chimaeric VLPs induced smaller tumour growth and decreased the number of metastases in the lung compared to wild-type VLPs. chimaeric VLPs induced high antibody titres towards the MUC1 epitope, as well as specific cellular responses towards P53 epitopes in lymph nodes local to the tumour. Our results reinforce and extend the utility of VP2 B19-VLPs as an encouraging tumour antigen delivery system in cancer immunotherapy able to improve tumour immunity in TNBC by inducing cellular and humoral immune responses.


Subject(s)
Antigens, Neoplasm/immunology , Cancer Vaccines/immunology , Parvovirus B19, Human/immunology , Triple Negative Breast Neoplasms/therapy , Vaccines, Virus-Like Particle/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Antigens, Neoplasm/administration & dosage , Bacillus thuringiensis Toxins/administration & dosage , Cancer Vaccines/administration & dosage , Cell Line, Tumor , Disease Models, Animal , Endotoxins/administration & dosage , Female , Hemolysin Proteins/administration & dosage , Humans , Immunity, Cellular , Immunity, Humoral , Immunogenicity, Vaccine , Insect Proteins , Mice , Receptors, Cell Surface , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/pathology , Vaccines, Virus-Like Particle/administration & dosage
2.
Dev Comp Immunol ; 121: 104071, 2021 08.
Article in English | MEDLINE | ID: mdl-33766585

ABSTRACT

The insecticidal Bacillus thuringiensis protein Cry1Ac is produced as a protoxin and becomes activated to a toxin when ingested by larvae. Both proteins are immunogenic and able to activate macrophages. The proposed mechanism of immunostimulation by Cry1Ac protoxin has been related to its capacity to activate antigen-presenting cells (APC), but its ability to activate dendritic cells (DC) has not been explored. Here we evaluated, in the popliteal lymph nodes (PLN), spleen and peritoneum, the activation of DC CD11c+ MHC-II+ following injection with single doses (50 µg) of Cry1Ac toxin or protoxin via the intradermal (i.d.) and intraperitoneal (i.p.) routes in C57BL/6 mice. In vivo stimulation with both Cry1Ac proteins induced activation of DC via upregulation of CD86, primarily in PLN 24 h after i. d. injection. Moreover, this activation was detected in DC, displaying CD103+, a typical marker of migratory DC, while upregulation of CD80 was uniquely induced by toxin. Tracking experiments showed that Cy5-labeled Cry1Ac proteins could rapidly reach the PLN and localize near DC, but some label remained in the footpad. When the capacity of Cry1Ac-activated DC to induce antigen presentation was examined, significant proliferation of naïve T lymphocytes was induced exclusively by the protoxin. The protoxin elicited a Th17-biased cytokine profile. Moreover, only the Cry1Ac toxin induced a pronounced proliferation of B cells from both untreated and Cry1Ac-injected mice, suggesting that it acts as a polyclonal activator. In conclusion, Cry1Ac protoxin and toxin show a distinctive capacity to activate APCs.


Subject(s)
B-Lymphocytes/immunology , Bacillus thuringiensis Toxins/immunology , Bacillus thuringiensis/immunology , Dendritic Cells/immunology , Endotoxins/immunology , Hemolysin Proteins/immunology , Animals , Antigen Presentation , B-Lymphocytes/metabolism , Bacillus thuringiensis Toxins/administration & dosage , Dendritic Cells/metabolism , Endotoxins/administration & dosage , Female , Hemolysin Proteins/administration & dosage , Lymphocyte Activation , Mice , Recombinant Proteins/administration & dosage , Recombinant Proteins/immunology
3.
Carbohydr Polym ; 254: 117312, 2021 Feb 15.
Article in English | MEDLINE | ID: mdl-33357875

ABSTRACT

Vitexin of Ficus deltoidea exhibits intestinal α-glucosidase inhibitory and blood glucose lowering effects. This study designs oral intestinal-specific alginate nanoparticulate system of vitexin. Nanospray-dried alginate, alginate/stearic acid and alginate-C18 conjugate nanoparticles were prepared. Stearic acid was adopted to hydrophobize the matrix and minimize premature vitexin release in stomach, whereas C-18 conjugate as immobilized fatty acid to sustain hydrophobic effect and drug release. Nanoparticles were compacted with polyethylene glycol (PEG 3000, 10,000 and 20,000). The physicochemical, drug release, in vivo blood glucose lowering and intestinal vitexin content of nanoparticles and compact were determined. Hydrophobization of alginate nanoparticles promoted premature vitexin release. Compaction of nanoparticles with PEG minimized vitexin release in the stomach, with stearic acid loaded nanoparticles exhibiting a higher vitexin release in the intestine. The introduction of stearic acid reduced vitexin-alginate interaction, conferred alginate-stearic acid mismatch, and dispersive stearic acid-induced particle breakdown with intestinal vitexin release. Use of PEG 10,000 in compaction brought about PEG-nanoparticles interaction that negated initial vitexin release. The PEG dissolution in intestinal phase subsequently enabled particle breakdown and vitexin release. The PEG compacted nanoparticles exhibited oral intestinal-specific vitexin release, with positive blood glucose lowering and enhanced intestinal vitexin content in vivo.


Subject(s)
Alginates/chemistry , Apigenin/administration & dosage , Bacterial Proteins/administration & dosage , Bacterial Toxins/administration & dosage , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/drug therapy , Drug Carriers/chemistry , Glycoside Hydrolase Inhibitors/administration & dosage , Hemolysin Proteins/administration & dosage , Hypoglycemic Agents/administration & dosage , Nanoparticles/chemistry , Administration, Oral , Alginates/metabolism , Animals , Apigenin/chemistry , Bacterial Proteins/chemistry , Bacterial Toxins/chemistry , Diabetes Mellitus, Experimental/chemically induced , Drug Liberation , Ficus/chemistry , Glycoside Hydrolase Inhibitors/chemistry , Hemolysin Proteins/chemistry , Hydrogen Bonding , Hypoglycemic Agents/chemistry , Male , Particle Size , Polyethylene Glycols/metabolism , Rats , Rats, Sprague-Dawley , Stearic Acids/chemistry , Streptozocin/adverse effects , alpha-Glucosidases/metabolism
4.
J Breath Res ; 15(1): 016003, 2020 10 24.
Article in English | MEDLINE | ID: mdl-33103661

ABSTRACT

Inflammation may alter volatile organic compounds (VOCs) in exhaled breath. We therefore used ion mobility spectrometry (IMS) to evaluate exhaled breath components in two non-infectious inflammatory models. Fifty male Sprague Dawley rats were anesthetized and ventilated for 24 h. Five treatments were randomly assigned: (1) lipopolysaccharide low dose [5 mg/kg]; (2) lipopolysaccharide high dose [10 mg/kg]; (3) alpha toxin low dose [40 µg/kg]; (4) alpha toxin high dose [80 µg/kg]; and, (5) NaCl 0.9% as control group. Gas was sampled from the expiratory line of the ventilator every 20 min and analyzed with IMS combined with a multi-capillary column. VOCs were identified by comparison with an established database. Survival analysis was performed by log-rank test, other analyses by one-way or paired ANOVA-tests and post-hoc analysis according to Holm-Sidak. Rats given NaCl and low-dose alpha toxin survived 24 h. The median survival time in alpha toxin high-dose group was 23 (95%-confidence interval (CI): 21, 24) h. In contrast, the median survival time in rats given high-dose lipopolysaccharide was 12 (95% CI: 9, 14) and only 13 (95% CI: 10, 16) h in those given high-dose lipopolysaccharide. 73 different VOCs were detected, of which 35 were observed only in the rats, 38 could be found both in the blank measurements of ventilator air and in the exhaled air of the rats. Forty-nine of the VOCs were identifiable from a registry of compounds. Exhaled volatile compounds were comparable in each group before injection of lipopolysaccharide and alpha toxin. In the LPS groups, 1-pentanol increased and 2-propanol decreased. After alpha toxin treatment, 1-butanol and 1-pentanol increased whereas butanal and isopropylamine decreased. Induction of a non-infectious systemic inflammation (niSI) by lipopolysaccharide and alpha toxin changes VOCs in exhaled breath. Exhalome analysis may help identify niSI.


Subject(s)
Bacterial Toxins/administration & dosage , Hemolysin Proteins/administration & dosage , Inflammation/pathology , Pulmonary Ventilation , Volatile Organic Compounds/analysis , Animals , Blood Chemical Analysis , Breath Tests , Cytokines/blood , Exhalation , Hemodynamics , Kaplan-Meier Estimate , Lipopolysaccharides/administration & dosage , Male , Rats, Sprague-Dawley , Survival Analysis
5.
J Gastroenterol Hepatol ; 35(12): 2241-2247, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32386240

ABSTRACT

BACKGROUND AND AIM: Mitochondrial damage is commonly involved in liver injury. We have previously shown that normal mitochondria can be coated with a carrier protein to form complexes that are specifically taken up by liver cells in culture. The aim of the current study was to determine whether mitochondrial complexes could be specifically delivered to the livers of living rats by intravenous injection. METHODS: Mitochondria were harvested from fresh mouse liver, mixed with an asialoglycoprotein-based carrier, asialoorosomucoid-polylysine (AsOR-PL), and purified to form complexes. To facilitate the release of internalized mitochondria from endosomes, an endosomolytic peptide, listeriolysin O (LLO), was coupled to AsOR to form AsOR-LLO. Mitochondria alone, mitochondrial complexes with AsOR-PL, and mitochondrial complexes plus AsOR-LLO conjugate all containing the same number of mitochondria were injected intravenously. Animals were killed, and organs were removed and analyzed by quantitative polymerase chain reaction of mouse mitochondrial DNA, electron microscopy (EM), and in situ polymerase chain reaction and hybridization followed by immunohistochemical analyses. RESULTS: Calculations revealed that approximately 27% of the total injected mitochondria was detected in the liver, while less than 2% was found in spleen, and < 1% in lungs. Immunohistochemistry showed that mouse mitochondrial DNA staining was minimal with mitochondrial complexes alone, strong periportal with mitochondrial complexes co-injected with AsOR-LLO, and absent with mitochondria alone. CONCLUSIONS: Targetable mitochondrial complexes can be delivered to rat liver, and the efficiency of that process is greatly enhanced by co-injection of a targetable endosomal release agent, AsOR-LLO.


Subject(s)
Asialoglycoproteins/administration & dosage , Bacterial Toxins/administration & dosage , Cell Transplantation/methods , Heat-Shock Proteins/administration & dosage , Hemolysin Proteins/administration & dosage , Liver , Mitochondria, Liver/transplantation , Orosomucoid/analogs & derivatives , Polylysine/administration & dosage , Animals , Carrier Proteins , Endosomes , Female , Hepatocytes/cytology , Injections, Intravenous , Mice, Inbred Strains , Orosomucoid/administration & dosage , Rats, Sprague-Dawley
6.
J Infect Dis ; 221(2): 267-275, 2020 01 02.
Article in English | MEDLINE | ID: mdl-31504652

ABSTRACT

Staphylococcus aureus is a common pathogen causing infections in humans with various degrees of severity, with pneumonia being one of the most severe infections. In as much as staphylococcal pneumonia is a disease driven in large part by α-hemolysin (Hla) and Panton-Valentine leukocidin (PVL), we evaluated whether active immunization with attenuated forms of Hla (HlaH35L/H48L) alone, PVL components (LukS-PVT28F/K97A/S209A and LukF-PVK102A) alone, or combination of all 3 toxoids could prevent lethal challenge in a rabbit model of necrotizing pneumonia caused by the USA300 community-associated methicillin-resistant S. aureus (MRSA). Rabbits vaccinated with Hla toxoid alone or PVL components alone were only partially protected against lethal pneumonia, whereas those vaccinated with all 3 toxoids had 100% protection against lethality. Vaccine-mediated protection correlated with induction of polyclonal antibody response that neutralized not only α-hemolysin and PVL, but also other related toxins, produced by USA300 and other epidemic MRSA clones.


Subject(s)
Bacterial Toxins/immunology , Exotoxins/immunology , Hemolysin Proteins/immunology , Leukocidins/immunology , Pneumonia, Necrotizing/prevention & control , Pneumonia, Staphylococcal/prevention & control , Animals , Bacterial Toxins/administration & dosage , Disease Models, Animal , Exotoxins/administration & dosage , Hemolysin Proteins/administration & dosage , Humans , Leukocidins/administration & dosage , Methicillin-Resistant Staphylococcus aureus , Pneumonia, Necrotizing/immunology , Pneumonia, Staphylococcal/immunology , Rabbits , Vaccination
7.
Insect Biochem Mol Biol ; 118: 103306, 2020 03.
Article in English | MEDLINE | ID: mdl-31843687

ABSTRACT

The insecticidal Cry toxins produced by Bacillus thuringiensis (Bt) are powerful tools for insect control. Cry toxin receptors such as cadherin (CAD), ABCC2 transporter and alkaline phosphatase (ALP), located on insect midgut cells, are needed for Cry toxicity. Although insect cell lines are useful experimental models for elucidating toxin action mechanism, most of them show low expression of Cry-receptors genes. The GATA transcription factor family plays important roles in regulating development and differentiation of intestine stem cells. Here, we investigated whether GATAs transcription factors are involved in the expression of Cry1Ac-receptors genes, using multiple insect cell lines. Four GATA genes were identified in the transcriptome of the midgut tissue from the lepidopteran larvae Helicoverpa armigera. These HaGATA genes were transiently expressed in three lepidopteran cell lines, Spodoptera frugiperda Sf9, H. armigera QB-Ha-E5 and Trichoplusia ni Hi5. Analysis of transcription activity using transcriptional gene-fusions showed that only H. armigera GATAe (HaGATAe) significantly increased the transcription of CAD, ABCC2 and ALP receptors genes in all insect cell lines. Key DNA regions for HaGATAe regulation were identified in the promoter sequence of these Cry-receptors genes by using promoter deletion mapping. The transient expression of HaGATAe in these cell lines, conferred sensitivity to Cry1Ac toxin, although in Hi5 cells the susceptibility to Cry1Ac was lower than in other two cell lines. High sensitivity to Cry1Ac correlated with simultaneous transcription of ABCC2 and CAD genes in Sf9 and QB-Ha-E5 cells. Our results reveal that HaGATAe enhances transcription of several lepidopteran Cry1Ac receptor genes in cultured insect cells.


Subject(s)
Bacterial Proteins/administration & dosage , Endotoxins/administration & dosage , GATA Transcription Factors/genetics , Gene Expression Regulation , Hemolysin Proteins/administration & dosage , Insect Proteins/genetics , Insecticide Resistance/genetics , Moths/genetics , Receptors, Cell Surface/genetics , Animals , Bacillus thuringiensis/chemistry , Bacillus thuringiensis Toxins , Cell Line , GATA Transcription Factors/metabolism , Insect Proteins/metabolism , Moths/drug effects , Moths/metabolism , Receptors, Cell Surface/metabolism , Sf9 Cells , Spodoptera/drug effects , Spodoptera/genetics , Spodoptera/metabolism
8.
Transgenic Res ; 29(1): 109-124, 2020 02.
Article in English | MEDLINE | ID: mdl-31781961

ABSTRACT

Safety assessment of genetically modified plants includes protein characterization to confirm the intended trait protein expression. In addition, to conduct safety tests, the large amount of purified protein needed is usually met through the use of a surrogate, microbially produced protein source. Characterization of the eCry3.1Ab and mCry3A proteins as derived from Event MZIR098 maize was challenging because of the difficulty in purifying/isolating these proteins that are of similar molecular weight and have considerable shared sequence and immunogenicity. This also applies to establishing the biochemical equivalence to the microbially produced surrogate proteins, as highly-purified plant protein is required. While use of crude plant extracts facilitated functional equivalence testing with the surrogate proteins, a separate technical challenge had to be met. The eCry3.1Ab and mCry3A proteins display differentiated modes of action toward CRW pests, however, with the same overall target pest spectrum, no differential test organism existed to allow equivalence testing for one insecticidal protein in the presence of the other. To establish that the microbially produced proteins are suitable surrogates for the plant-produced proteins, the challenges in the protein purification and bioactivity testing had to be addressed. This article describes technical solutions to assess and characterize the insecticidal proteins in this new event and thereby confirm equivalence/suitability of the microbially produced protein surrogates.


Subject(s)
Bacillus thuringiensis Toxins/administration & dosage , Bacillus thuringiensis/metabolism , Coleoptera/drug effects , Endotoxins/administration & dosage , Hemolysin Proteins/administration & dosage , Plants, Genetically Modified/metabolism , Zea mays/metabolism , Amino Acid Sequence , Animals , Bacillus thuringiensis/genetics , Bacillus thuringiensis Toxins/metabolism , Endotoxins/metabolism , Glycosylation , Hemolysin Proteins/metabolism , Plants, Genetically Modified/genetics , Zea mays/genetics
9.
Am J Trop Med Hyg ; 101(5): 1177-1182, 2019 11.
Article in English | MEDLINE | ID: mdl-31516117

ABSTRACT

Strongyloidiasis, caused by Strongyloides stercoralis infection, is an important neglected tropical disease that causes significant public health problems in the tropics and subtropics. The disease can persist in hosts for decades and may be life-threatening because of hyperinfection and dissemination. Ivermectin (mostly) and albendazole are the most common anthelmintics used for treatment. Albendazole is suboptimal for this parasite, and although ivermectin is quite effective in immunocompromised patients, a multiple-course regimen is required. Furthermore, reliance on a single drug class for treating intestinal nematodes is a recipe for future failure. Therefore, it is important to discover new anthelmintics to treat or prevent human strongyloidiasis. One promising candidate is the Bacillus thuringiensis crystal protein Cry5B. Cry5B is highly potent against parasitic nematodes, for example, hookworms and Ascaris suum. Here, we investigated the potential of Cry5B against S. stercoralis. Multiple stages of S. stercoralis, including the first larval stage (L1s), infective stage (iL3s), free-living adult stage, and parasitic female stage, were all susceptible to Cry5B as indicated by impairment of motility and decreased viability in vitro. In summary, Cry5B demonstrated strong potential as an effective anthelmintic for treatment and transmission control of human strongyloidiasis, justifying further experiments to investigate in vivo therapeutic efficacy.


Subject(s)
Bacterial Proteins/pharmacology , Endotoxins/pharmacology , Hemolysin Proteins/pharmacology , Strongyloides stercoralis/drug effects , Albendazole/pharmacology , Animals , Anthelmintics/administration & dosage , Anthelmintics/pharmacology , Bacillus thuringiensis Toxins , Bacterial Proteins/administration & dosage , Dose-Response Relationship, Drug , Endotoxins/administration & dosage , Escherichia coli/classification , Escherichia coli/metabolism , Female , Hemolysin Proteins/administration & dosage , Ivermectin/pharmacology , Larva/drug effects , Recombinant Proteins/pharmacology
10.
Sci Rep ; 9(1): 9661, 2019 07 04.
Article in English | MEDLINE | ID: mdl-31273223

ABSTRACT

Parasporal inclusions of a native non haemolytic Bacillus thuringiensis strain KAU 59 was screened for its cytotoxicity against human lymphocytic leukemic cell line jurkat and normal human lymphocytes. The cytotoxicity of proteinase activated and non activated solubilised parasporal inclusions against both cell lines was assessed by Cell Titer 96 Aqueous Non Radioactive Cell Proliferation Assay Kit using MTS. The 50 per cent effective concentration (EC50) values were deduced from log probit analysis at 48 h. Morphological changes associated with cytotoxicity were evaluated and molecular mechanisms of cell death were elucidated by TUNEL assay at 48 h post-inoculation. The fluorescence assisted cell sorting was done in the flow cytometer to assess the stage of cell cycle arrest. Relative quantification of caspase-3 expression in Jurkat cells treated with parasporal inclusion protein of KAU 59 was done by qRTPCR The results indicated that the protein was cytotoxic to jurkat cells at the same time non toxic to normal lymphocytes. Cytotoxicity was evident only after proteolytic activation. Apoptotic cell death was confirmed in the protein treated cells by TUNEL Assay and also up regulated caspase-3 gene expression (P < 0.001). S phase cell cycle arrest was confirmed by and fluorescence associated cell sorting.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis , Bacterial Proteins/administration & dosage , Cell Cycle Checkpoints , Endotoxins/administration & dosage , Hemolysin Proteins/administration & dosage , Hemolysis/drug effects , Leukemia/pathology , Bacillus thuringiensis Toxins , Humans , Leukemia/drug therapy , Leukemia/metabolism , Tumor Cells, Cultured
11.
Toxicon ; 164: 82-86, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30991063

ABSTRACT

Conventional drug delivery systems have many limitations including cytotoxicity and affecting non-specific cells. Cell-targeting peptides (CTPs) as a potential class of targeting moiety have some advantages over previous targeting moieties such as monoclonal antibodies, offer additional benefits to design systems using CTPs. Here we have engineered listeriolysin O (LLO) pore-forming toxin by adding a luteinizing hormone-releasing hormone (LHRH) targeting peptide to its N-terminus. Two versions of the toxin, with and without targeting peptide, were sub-cloned into a bacterial expression plasmid. BL21 DE3 cells were used for induction of expression and recombinant proteins were purified using nickel-immobilized metal affinity chromatography column. In order to treat MDA-MB-231 and SKOV3 cell lines as LHRH receptor positive and negative cells, two mentioned LLO toxins were used to evaluate their cytotoxicity and specificity. Our results reveal that the IC50 of LLO toxin on MDA-MB-231 and SKOV3 cells was 0.32 and 0.41 µg/ml respectively. Furthermore, IC50 of fusion LHRH-LLO toxin on the cells was 0.88 and 19.55 µg/ml. Cytotoxicity of engineered LHRH-LLO toxin on negative cells was significantly 48-fold lower than wild-type LLO toxin. But this difference has been lowered to only 2.7-fold less cytotoxicity in positive cells. To the best of our knowledge, the current work as the first study regarding engineered toxin revealed that CDC family members could be used to target the specific cell-type.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Bacterial Toxins/pharmacokinetics , Drug Delivery Systems/methods , Gonadotropin-Releasing Hormone/pharmacokinetics , Heat-Shock Proteins/pharmacokinetics , Hemolysin Proteins/pharmacokinetics , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Bacterial Toxins/administration & dosage , Bacterial Toxins/pharmacology , Cell Line, Tumor , Erythrocytes/drug effects , Escherichia coli/metabolism , Heat-Shock Proteins/administration & dosage , Heat-Shock Proteins/pharmacology , Hemolysin Proteins/administration & dosage , Hemolysin Proteins/pharmacology , Hemolysis , Humans , Molecular Structure , Receptors, LHRH/metabolism , Recombinant Proteins
12.
J Appl Microbiol ; 125(4): 997-1007, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29877008

ABSTRACT

AIMS: The aim of this study was to screen vaccine candidates from virulence factors of Streptococcus iniae in flounder model. METHODS AND RESULTS: The immunogenicity of recombinant phosphoglucomutase (rPGM) and rCAMP factor was confirmed by Western blot. The percentage of surface membrane immunoglobulin-positive (sIg+ ) lymphocytes in peripheral blood leucocytes, the specific and total serum IgM and the activity of acid phosphatase (ACP) and peroxidase (POD) in flounder were determined with flow cytometry, ELISA and commercial enzyme activity kits, respectively, after intraperitoneal immunization with rPGM and rCAMP factor. The results showed that rPGM and rCAMP factor could induce significant rise in sIg+ lymphocytes, specific serum IgM and activities of ACP and POD. Additionally, the relative percent survival rate of the vaccinated flounder was 64 and 54% in challenge experiment using S. iniae, respectively. These results indicated that rPGM and rCAMP factor could evoke humoural and innate immune response in flounder and provide high-efficiency immunoprotection against S. iniae infection. CONCLUSIONS: Phosphoglucomutase (PGM) and CAMP factor were promising vaccine candidates against S. iniae in flounder. SIGNIFICANCE AND IMPACT OF THE STUDY: Phosphoglucomutase and CAMP factor have the potential to be vaccine candidates, which provide important information for us to develop the effective subunit vaccines, especially the multivaccine, against S .iniae in aquaculture.


Subject(s)
Bacterial Proteins/immunology , Bacterial Vaccines/immunology , Fish Diseases/prevention & control , Hemolysin Proteins/immunology , Phosphoglucomutase/immunology , Streptococcal Infections/veterinary , Streptococcus iniae/immunology , Vaccines, Subunit/immunology , Animals , Bacterial Proteins/administration & dosage , Bacterial Proteins/genetics , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/genetics , Fish Diseases/immunology , Fish Diseases/microbiology , Flounder/microbiology , Hemolysin Proteins/administration & dosage , Hemolysin Proteins/genetics , Immunity, Innate , Phosphoglucomutase/administration & dosage , Phosphoglucomutase/genetics , Streptococcal Infections/immunology , Streptococcal Infections/microbiology , Streptococcal Infections/prevention & control , Streptococcus iniae/enzymology , Streptococcus iniae/genetics , Vaccination , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/genetics
13.
Int J Parasitol Drugs Drug Resist ; 8(2): 287-294, 2018 08.
Article in English | MEDLINE | ID: mdl-29772478

ABSTRACT

Hookworms are intestinal nematode parasites that infect nearly half a billion people and are globally one of the most important contributors to iron-deficiency anemia. These parasites have significant impacts in developing children, pregnant women and working adults. Of all the soil-transmitted helminths or nematodes (STNs), hookworms are by far the most important, with disease burdens conservatively estimated at four million DALYs (Disability-Adjusted Life Years) and with productivity losses of up to US$139 billion annually. To date, mainly one drug, albendazole is used for hookworm therapy in mass drug administration, which has on average ∼80% cure rate that is lower (<40%) in some places. Given the massive numbers of people needing treatment, the threat of parasite resistance, and the inadequacy of current treatments, new and better cures against hookworms are urgently needed. Cry5B, a pore-forming protein produced by the soil bacterium Bacillus thuringiensis (Bt) has demonstrated good efficacy against Ancylostoma ceylanicum hookworm infections in hamsters. Here we broaden studies of Cry5B to include tests against infections of Ancylostoma caninum hookworms in dogs and against infections of the dominant human hookworm, Necator americanus, in hamsters. We show that Cry5B is highly effective against all hookworm parasites tested in all models. Neutralization of stomach acid improves Cry5B efficacy, which will aid in practical application of Cry5B significantly. Importantly, we also demonstrate that the anti-nematode therapeutic efficacy of Cry5B is independent of the host immune system and is not itself negated by repeated dosing. This study indicates that Bt Cry5B is a pan-hookworm anthelmintic with excellent properties for use in humans and other animals.


Subject(s)
Ancylostomatoidea/drug effects , Anthelmintics/therapeutic use , Bacillus thuringiensis/chemistry , Bacterial Proteins/therapeutic use , Endotoxins/therapeutic use , Hemolysin Proteins/therapeutic use , Hookworm Infections/drug therapy , Ancylostoma/drug effects , Ancylostomiasis/drug therapy , Ancylostomiasis/parasitology , Animals , Anthelmintics/administration & dosage , Bacillus thuringiensis Toxins , Bacterial Proteins/administration & dosage , Cricetinae , Dogs , Endotoxins/administration & dosage , Hemolysin Proteins/administration & dosage , Intestinal Diseases, Parasitic/drug therapy , Necator americanus/drug effects , Necatoriasis/drug therapy , Necatoriasis/parasitology
14.
PLoS Negl Trop Dis ; 12(5): e0006506, 2018 05.
Article in English | MEDLINE | ID: mdl-29775454

ABSTRACT

BACKGROUND: The soil-transmitted nematodes (STNs) or helminths (hookworms, whipworms, large roundworms) infect the intestines of ~1.5 billion of the poorest peoples and are leading causes of morbidity worldwide. Only one class of anthelmintic or anti-nematode drugs, the benzimidazoles, is currently used in mass drug administrations, which is a dangerous situation. New anti-nematode drugs are urgently needed. Bacillus thuringiensis crystal protein Cry5B is a powerful, promising new candidate. Drug combinations, when properly made, are ideal for treating infectious diseases. Although there are some clinical trials using drug combinations against STNs, little quantitative and systemic work has been performed to define the characteristics of these combinations in vivo. METHODOLOGY/PRINCIPAL FINDINGS: Working with the hookworm Ancylostoma ceylanicum-hamster infection system, we establish a laboratory paradigm for studying anti-nematode combinations in vivo using Cry5B and the nicotinic acetylcholine receptor (nAChR) agonists tribendimidine and pyrantel pamoate. We demonstrate that Cry5B strongly synergizes in vivo with both tribendimidine and pyrantel at specific dose ratios against hookworm infections. For example, whereas 1 mg/kg Cry5B and 1 mg/kg tribendimidine individually resulted in only a 0%-6% reduction in hookworm burdens, the combination of the two resulted in a 41% reduction (P = 0.020). Furthermore, when mixed at synergistic ratios, these combinations eradicate hookworm infections at doses where the individual doses do not. Using cyathostomin nematode parasites of horses, we find based on inhibitory concentration 50% values that a strongylid parasite population doubly resistant to nAChR agonists and benzimidazoles is more susceptible or "hypersusceptible" to Cry5B than a cyathostomin population not resistant to nAChR agonists, consistent with previous Caenhorhabditis elegans results. CONCLUSIONS/SIGNIFICANCE: Our study provides a powerful means by which anthelmintic combination therapies can be examined in vivo in the laboratory. In addition, we demonstrate that Cry5B and nAChR agonists have excellent combinatorial properties-Cry5B combined with nAChR agonists gives rise to potent cures that are predicted to be recalcitrant to the development of parasite resistance. These drug combinations highlight bright spots in new anthelmintic development for human and veterinary animal intestinal nematode infections.


Subject(s)
Ancylostoma/drug effects , Ancylostomiasis/drug therapy , Ancylostomiasis/veterinary , Anthelmintics/administration & dosage , Bacterial Proteins/administration & dosage , Endotoxins/administration & dosage , Hemolysin Proteins/administration & dosage , Intestinal Diseases, Parasitic/drug therapy , Intestinal Diseases, Parasitic/veterinary , Nicotinic Antagonists/administration & dosage , Ancylostoma/physiology , Ancylostomiasis/parasitology , Animals , Bacillus thuringiensis Toxins , Cricetinae , Drug Therapy, Combination , Female , Horse Diseases/drug therapy , Horse Diseases/parasitology , Horses , Humans , Intestinal Diseases, Parasitic/parasitology , Male , Mesocricetus , Receptors, Nicotinic/genetics , Receptors, Nicotinic/metabolism
15.
BMC Vet Res ; 12(1): 101, 2016 Jun 13.
Article in English | MEDLINE | ID: mdl-27297520

ABSTRACT

BACKGROUND: Bovine necrohemorrhagic enteritis is caused by Clostridium perfringens type A. Due to the rapid progress and fatal outcome of the disease, vaccination would be of high value. In this study, C. perfringens toxins, either as native toxins or after formaldehyde inactivation, were evaluated as possible vaccine antigens. We determined whether antisera raised in calves against these toxins were able to protect against C. perfringens challenge in an intestinal loop model for bovine necrohemorrhagic enteritis. RESULTS: Alpha toxin and perfringolysin O were identified as the most immunogenic proteins in the vaccine preparations. All vaccines evoked a high antibody response against the causative toxins, alpha toxin and perfringolysin O, as detected by ELISA. All antibodies were able to inhibit the activity of alpha toxin and perfringolysin O in vitro. However, the antibodies raised against the native toxins were more inhibitory to the C. perfringens-induced cytotoxicity (as tested on bovine endothelial cells) and only these antibodies protected against C. perfringens challenge in the intestinal loop model. CONCLUSION: Although immunization of calves with both native and formaldehyde inactivated toxins resulted in high antibody titers against alpha toxin and perfringolysin O, only antibodies raised against native toxins protect against C. perfringens challenge in an intestinal loop model for bovine necrohemorrhagic enteritis.


Subject(s)
Antibodies, Neutralizing/immunology , Bacterial Toxins/administration & dosage , Bacterial Vaccines/administration & dosage , Calcium-Binding Proteins/administration & dosage , Cattle Diseases/microbiology , Clostridium Infections/veterinary , Clostridium perfringens/immunology , Enteritis/veterinary , Hemolysin Proteins/administration & dosage , Type C Phospholipases/administration & dosage , Animals , Bacterial Toxins/immunology , Bacterial Toxins/toxicity , Bacterial Vaccines/immunology , Bacterial Vaccines/toxicity , Calcium-Binding Proteins/immunology , Calcium-Binding Proteins/toxicity , Cattle , Cattle Diseases/immunology , Cattle Diseases/prevention & control , Clostridium Infections/immunology , Clostridium Infections/pathology , Clostridium Infections/prevention & control , Disease Models, Animal , Endothelial Cells/immunology , Enteritis/immunology , Enteritis/pathology , Enteritis/prevention & control , Hemolysin Proteins/immunology , Hemolysin Proteins/toxicity , Jejunum/immunology , Male , Necrosis , Type C Phospholipases/immunology , Type C Phospholipases/toxicity
16.
J Control Release ; 233: 114-25, 2016 07 10.
Article in English | MEDLINE | ID: mdl-27189136

ABSTRACT

The development of subunit mucosal vaccines requires an appropriate delivery system or an immune modulator such as an adjuvant to improve antigen immunogenicity. The nasal route for vaccine delivery by microparticles has attracted considerable interest, although challenges such as the rapid mucociliary clearance in the respiratory mucosa and the low immunogenicity of subunit vaccine still remain. Here, we aimed to develop mannan-decorated mucoadhesive thiolated hydroxypropylmethyl cellulose phthalate (HPMCP) microspheres (Man-THM) that contain ApxIIA subunit vaccine - an exotoxin fragment as a candidate for a subunit nasal vaccine against Actinobacillus pleuropneumoniae. For adjuvant activity, mucoadhesive thiolated HPMCP microspheres decorated with mannan could be targeted to the PRRs (pathogen recognition receptors) and mannose receptors (MR) of antigen presenting cells (APCs) in the respiratory immune system. The potential adjuvant ability of Man-THM for intranasal immunization was confirmed by in vitro and in vivo experiments. In a mechanistic study using APCs in vitro, it was found that Man-THM enhanced receptor-mediated endocytosis by stimulating the MR of APCs. In vivo, the nasal vaccination of ApxIIA-loaded Man-THM in mice resulted in higher levels of mucosal sIgA and serum IgG than mice in the ApxIIA and ApxIIA-loaded THM groups due to the specific recognition of the mannan in the Man-THM by the MRs of the APCs. Moreover, ApxIIA-containing Man-THM protected immunized mice when challenged with strains of A. pleuropneumoniae serotype 5. These results suggest that mucoadhesive Man-THM may be a promising candidate for a nasal vaccine delivery system to elicit systemic and mucosal immunity that can protect from pathogenic bacteria infection.


Subject(s)
Actinobacillus Infections/prevention & control , Bacterial Proteins/administration & dosage , Bacterial Vaccines/administration & dosage , Hemolysin Proteins/administration & dosage , Mannans/administration & dosage , Methylcellulose/analogs & derivatives , Actinobacillus pleuropneumoniae/immunology , Adhesiveness , Administration, Intranasal , Animals , Antibodies, Bacterial/blood , Antigen-Presenting Cells/immunology , Bacterial Proteins/chemistry , Bacterial Proteins/immunology , Bacterial Vaccines/chemistry , Bronchoalveolar Lavage Fluid/immunology , Cell Line , Female , Hemolysin Proteins/chemistry , Hemolysin Proteins/immunology , Immunity, Mucosal , Immunization/methods , Immunoglobulin A/blood , Immunoglobulin A/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Mannans/chemistry , Methylcellulose/administration & dosage , Methylcellulose/chemistry , Mice , Mice, Inbred BALB C , Microspheres , Respiratory Mucosa/chemistry , Sulfhydryl Compounds/administration & dosage , Sulfhydryl Compounds/chemistry
17.
Clin Vaccine Immunol ; 23(6): 442-50, 2016 06.
Article in English | MEDLINE | ID: mdl-27030589

ABSTRACT

Staphylococcus aureus alpha-hemolysin (Hla) assembles into heptameric pores on the host cell membrane, causing lysis, apoptosis, and junction disruption. Herein, we present the design of a newly engineered S. aureus alpha-toxin, HlaPSGS, which lacks the predicted membrane-spanning stem domain. This protein is able to form heptamers in aqueous solution in the absence of lipophilic substrata, and its structure, obtained by transmission electron microscopy and single-particle reconstruction analysis, resembles the cap of the wild-type cytolytic Hla pore. HlaPSGS was found to be impaired in binding to host cells and to its receptor ADAM10 and to lack hemolytic and cytotoxic activity. Immunological studies using human sera as well as sera from mice convalescent from S. aureus infection suggested that the heptameric conformation of HlaPSGS mimics epitopes exposed by the cytolytic Hla pore during infection. Finally, immunization with this newly engineered Hla generated high protective immunity against staphylococcal infection in mice. Overall, this study provides unprecedented data on the natural immune response against Hla and suggests that the heptameric HlaPSGS is a highly valuable vaccine candidate against S. aureus.


Subject(s)
Bacterial Toxins/chemistry , Bacterial Toxins/immunology , Hemolysin Proteins/chemistry , Hemolysin Proteins/immunology , Molecular Mimicry , Staphylococcal Infections/prevention & control , Staphylococcus aureus , ADAM10 Protein/metabolism , Animals , Bacterial Toxins/administration & dosage , Bacterial Toxins/genetics , Cell Line , Cytotoxins , Epitopes/immunology , Escherichia coli/genetics , Hemolysin Proteins/administration & dosage , Hemolysin Proteins/genetics , Humans , Membrane Proteins/metabolism , Mice , Microscopy, Electron, Transmission , Models, Molecular , Protein Engineering , Recombinant Proteins/administration & dosage , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/isolation & purification , Staphylococcal Vaccines/immunology , Staphylococcus aureus/chemistry , Staphylococcus aureus/metabolism , Vaccination
18.
Sci Rep ; 6: 20368, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26829252

ABSTRACT

Plant varieties expressing the Bt (Bacillus thuringiensis) insecticidal proteins Cry1Ah and Cry2Ab have potential commercialization prospects in China. However, their potential effects on non-target arthropods (NTAs) remain uncharacterized. The cotton aphid Aphis gossypii is a worldwide pest that damages various important crops. The ladybeetle Propylea japonica is a common and abundant natural enemy in many cropping systems in East Asia. In the present study, the effects of Cry1Ah and Cry2Ab proteins on A. gossypii and P. japonica were assessed from three aspects. First, neither of the Cry proteins affected the growth or developmental characteristics of the two test insects. Second, the expression levels of the detoxification-related genes of the two test insects did not change significantly in either Cry protein treatment. Third, neither of the Cry proteins had a favourable effect on the expression of genes associated with the amino acid metabolism of A. gossypii and the nutrition utilization of P. japonica. In conclusion, the Cry1Ah and Cry2Ab proteins do not appear to affect the cotton aphid A. gossypii or the ladybeetle P. japonica.


Subject(s)
Aphids/drug effects , Aphids/physiology , Bacillus thuringiensis/metabolism , Bacterial Proteins/pharmacology , Coleoptera/drug effects , Coleoptera/physiology , Endotoxins/pharmacology , Hemolysin Proteins/pharmacology , Insect Control , Amino Acids/metabolism , Animals , Bacillus thuringiensis Toxins , Bacterial Proteins/administration & dosage , Endotoxins/administration & dosage , Gene Expression Regulation/drug effects , Genes, Insect , Gossypium/genetics , Gossypium/parasitology , Hemolysin Proteins/administration & dosage , Inactivation, Metabolic/genetics , Plants, Genetically Modified
19.
Transgenic Res ; 25(2): 163-72, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26581349

ABSTRACT

Numerous genetically modified (GM) crops expressing proteins for insect resistance have been commercialized following extensive testing demonstrating that the foods obtained from them are as safe as that obtained from their corresponding non-GM varieties. In this paper, we report the outcome of safety studies conducted on a newly developed insect-resistant GM rice expressing the cry2A* gene by a subchronic oral toxicity study on rats. GM rice and non-GM rice were incorporated into the diet at levels of 30, 50, and 70% (w/w), No treatment-related adverse or toxic effects were observed based on an examination of the daily clinical signs, body weight, food consumption, hematology, serum biochemistry, and organ weight or based on gross and histopathological examination. These results demonstrate that the GM rice with cry2A* gene is as safe for food as conventional non-GM rice.


Subject(s)
Bacterial Proteins/genetics , Endotoxins/genetics , Food Analysis , Hemolysin Proteins/genetics , Oryza/genetics , Plants, Genetically Modified/genetics , Animals , Bacillus thuringiensis Toxins , Bacterial Proteins/administration & dosage , Bacterial Proteins/adverse effects , Body Weight/genetics , Endotoxins/administration & dosage , Endotoxins/adverse effects , Hemolysin Proteins/administration & dosage , Hemolysin Proteins/adverse effects , Humans , Lepidoptera/genetics , Lepidoptera/pathogenicity , Oryza/adverse effects , Oryza/growth & development , Plants, Genetically Modified/adverse effects , Plants, Genetically Modified/growth & development , Rats
20.
Biochem Biophys Res Commun ; 469(3): 698-703, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26692482

ABSTRACT

Bacillus thuringiensis Cry4Ba mosquito-active toxin was previously shown to utilize two critical loop-residues, Tyr(332) and Phe(364) which are respectively located in ß2-ß3 and ß4-ß5 loops, for synergistic interactions with its alternative receptor-Cyt2Aa2. Here, structural analysis of the Cry4Ba-receptor-binding domain revealed that its N-terminal subdomain encompasses ß2-ß3 and ß4-ß5 hairpins which are stabilized by inter-hairpin hydrogen bonding between Thr(328) in ß2 and Thr(369) in ß5. Functional importance of these two side-chains was demonstrated by single-Ala substitutions (T328A and T369A), adversely affecting toxin activity against Aedes aegypti larvae. Unlike toxicity restoration of the inactive E417A/Y455A toxin mutated within another receptor-binding subdomain, defective bioactivity of T328A and T369A mutants cannot be restored by Cyt2Aa2 as also observed for ß2-ß3 (Y332A) and ß4-ß5 (F364A) loop-mutants. ELISA-based analysis further verified a loss in binding of all four bio-inactive mutants (T328A, Y332A, T369A and F364A) to the immobilized Cyt2Aa2. Protein-protein docking suggested that the two critical loop-residues (Tyr(332) and Phe(364)) correspondingly located at ß2-ß3 and ß4-ß5 loops can clearly interact with four counterpart surface-exposed residues of Cyt2Aa2. Altogether, our present data demonstrate structural importance of Thr(328) and Thr(369) toward hydrogen-bonded stabilization of two receptor-binding hairpins (ß2-ß3 and ß4-ß5) for synergistic toxicity of Cry4Ba with Cyt2Aa2.


Subject(s)
Aedes/drug effects , Bacterial Proteins/administration & dosage , Bacterial Proteins/chemistry , Endotoxins/administration & dosage , Endotoxins/chemistry , Hemolysin Proteins/administration & dosage , Hemolysin Proteins/chemistry , Models, Biological , Molecular Docking Simulation , Mosquito Control/methods , Aedes/physiology , Animals , Bacillus thuringiensis Toxins , Bacterial Proteins/ultrastructure , Binding Sites , Drug Synergism , Hemolysin Proteins/ultrastructure , Models, Chemical , Protein Binding , Protein Conformation , Structure-Activity Relationship , Survival Rate , Threonine/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...