Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 676
Filter
1.
Sci Rep ; 11(1): 18641, 2021 09 20.
Article in English | MEDLINE | ID: mdl-34545126

ABSTRACT

Human respiratory syncytial virus (hRSV) infection is a major pediatric health concern worldwide. Despite more than half a century of efforts, there is still no commercially available vaccine. In this study, we constructed and purified the recombinant protein CTA1-DD-RBF composed of a CTA1-DD mucosal adjuvant and prefusion F protein (RBF) using Escherichia coli BL21 cells. We studied the immunogenicity of CTA1-DD-RBF in mice. Intranasal immunization with CTA1-DD-RBF stimulated hRSV F-specific IgG1, IgG2a, sIgA, and neutralizing antibodies as well as T cell immunity without inducing lung immunopathology upon hRSV challenge. Moreover, the protective immunity of CTA1-DD-RBF was superior to that of the RBF protein, as confirmed by the assessment of serum-neutralizing activity and viral clearance after challenge. Compared to formalin-inactivated hRSV (FI-RSV), intranasal immunization with CTA1-DD-RBF induced a Th1 immune response. In summary, intranasal immunization with CTA1-DD-RBF is safe and effective in mice. Therefore, CTA1-DD-RBF represents a potential mucosal vaccine candidate for the prevention of human infection with hRSV.


Subject(s)
Cholera Toxin/administration & dosage , Cholera Toxin/immunology , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/immunology , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus, Human/immunology , Viral Fusion Proteins/administration & dosage , Viral Fusion Proteins/immunology , Adjuvants, Vaccine/administration & dosage , Administration, Intranasal , Animals , Antibodies, Neutralizing/biosynthesis , Antibodies, Viral/biosynthesis , Cholera Toxin/genetics , Female , Humans , Immunity, Mucosal , Immunization , Lung/immunology , Lung/pathology , Lung/virology , Mice , Mice, Inbred BALB C , Recombinant Fusion Proteins/genetics , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus, Human/genetics , Th1 Cells/immunology , Viral Fusion Proteins/genetics , Virus Replication
2.
Int J Mol Sci ; 22(13)2021 Jun 29.
Article in English | MEDLINE | ID: mdl-34209932

ABSTRACT

Enzymatic transamidation of gliadins by microbial transglutaminase (mTG) inhibits interferon-γ (IFN-γ) secretion by intestinal T cell lines in patients with celiac disease (CD). To gain insight into the cellular mechanisms underlying the down-regulatory effects of transamidation, we tested a single recombinant α-gliadin (r-gliadin) harbouring two immunodominant peptides, p13 (aa. 120-139) and p23 (aa. 220-239), in HLA-DQ8 transgenic mice, a model of gluten sensitivity. Mice were intranasally immunised with r-gliadin or r-gliadin transamidated by mTG (K-r-gliadin) along with cholera toxin, and the response of mesenteric lymph node cells was analysed by cytokine multiplex assay. An in vitro challenge with r-gliadin was characterised by secretion of specific cytokines featuring both innate immunity and the Th1/Th2/Th17 pattern of the adaptive response. Notably, transamidation specifically down-regulated the Th1 response. Structural studies performed on K-r-gliadin confirmed that specific glutamine residues in p13 and p23, previously found to be deamidated by tissue transglutaminase, were also transamidated by mTG. In silico analysis, simulating p13 and p23 peptide binding to HLA-DQ8 showed that these glutamines, in the form of glutamate, could interact by means of salt bridges with peculiar amino acids of the alpha chain of HLA-DQ8, suggesting that their transamidation may influence the HLA-restricted recognition of these peptides. Thus, the structural findings provided a rationale to explain the down-regulation of the r-gliadin-specific Th1 response following transamidation.


Subject(s)
Celiac Disease/drug therapy , Cholera Toxin/administration & dosage , Cytokines/metabolism , Gliadin/administration & dosage , HLA-DQ Antigens/genetics , Transglutaminases/metabolism , Administration, Intranasal , Animals , Celiac Disease/genetics , Celiac Disease/immunology , Cholera Toxin/immunology , Cytokines/drug effects , Disease Models, Animal , Down-Regulation , Gene Expression Regulation , Gliadin/chemistry , Gliadin/genetics , Gliadin/immunology , HLA-DQ Antigens/metabolism , Immunization , Immunodominant Epitopes/immunology , Mice , Mice, Transgenic , Recombinant Proteins/administration & dosage , Recombinant Proteins/chemistry , Recombinant Proteins/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Th2 Cells/immunology
3.
Methods Mol Biol ; 2223: 49-65, 2021.
Article in English | MEDLINE | ID: mdl-33226586

ABSTRACT

Egg allergy is one of the most common food allergies in children, being the most important allergenic proteins found in the egg white (EW). Allergy to EW shows a complex phenotype that involves a multifaceted reaction that can only be assessed in vivo. Although other routes of sensitization have been described, oral exposure to food antigens is one of the most suitable in humans. In mice, oral administration of allergenic proteins results in the development of tolerance, and the use of adjuvants, such as cholera toxin (CT), is required to promote Th2-biased immune responses over tolerogenic responses. In this regard, among the mouse strains that readily display Th2 responses, Balb/c has been widely used. Here, we describe a frequently used protocol of oral EW sensitization by using CT as an adjuvant and we explain in detail the methods that we have developed to analyze the sensitizing and eliciting capacity of EW proteins including evaluation of signs, measurement of serum levels of specific immunoglobulins, mast cell degranulation, cytokine secretion profile of allergen-reactive T cells, phenotyping of mesenteric lymph node- and spleen-derived dendritic and T cells by flow cytometry, and quantification of intestinal gene expression.


Subject(s)
Dendritic Cells/drug effects , Disease Models, Animal , Egg Hypersensitivity/immunology , Egg White/chemistry , Immunophenotyping/methods , Th2 Cells/drug effects , Adjuvants, Immunologic/administration & dosage , Administration, Oral , Animals , Biomarkers/metabolism , Chemokine CCL2/genetics , Chemokine CCL2/immunology , Chickens , Cholera Toxin/administration & dosage , Dendritic Cells/cytology , Dendritic Cells/immunology , Egg Hypersensitivity/blood , Egg Hypersensitivity/genetics , Egg Hypersensitivity/pathology , Female , Flow Cytometry , Gene Expression , Humans , Immunoglobulins/blood , Immunoglobulins/classification , Immunoglobulins/immunology , Interleukins/genetics , Interleukins/immunology , Lymph Nodes/cytology , Lymph Nodes/drug effects , Lymph Nodes/immunology , Mice , Mice, Inbred BALB C , Spleen/cytology , Spleen/drug effects , Spleen/immunology , Th2 Cells/cytology , Th2 Cells/immunology
4.
Methods Mol Biol ; 2223: 67-78, 2021.
Article in English | MEDLINE | ID: mdl-33226587

ABSTRACT

Cow's milk allergy is one of the most prevalent food allergies in both children and adults. As dairy products are common dietary ingredients and the prevalence of chronic conditions is on the rise, milk allergy is a growing public health concern. To elucidate underlying mechanisms and develop therapeutic strategies, reliable animal models are essential research tools. Sensitization to a milk protein is the principal procedure for establishing animal models of cow's milk allergy. However, the methods of sensitization vary from laboratory to laboratory, using different milk proteins with different amounts, routes, and durations of allergen exposure during sensitization of varying sex and strains of mice, likely resulting in diverse immunological and physical responses. Furthermore, the sources and potential impurities of milk protein may also produce variable responses. Thus, standardization of sensitization protocol is important, particularly when results are compared across studies. Here, we describe a method to generate a mouse model of cow's milk allergy using purified ß-lactoglobulin as the milk allergen with cholera toxin as an adjuvant in a 5-week oral sensitization protocol.


Subject(s)
Anaphylaxis/immunology , Disease Models, Animal , Lactoglobulins/immunology , Milk Hypersensitivity/immunology , Milk/immunology , Adjuvants, Immunologic/administration & dosage , Administration, Oral , Allergens/administration & dosage , Allergens/immunology , Anaphylaxis/blood , Anaphylaxis/pathology , Animals , Cattle , Cholera Toxin/administration & dosage , Female , Humans , Immunoglobulin E/blood , Immunoglobulin E/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Lactoglobulins/administration & dosage , Male , Mice , Mice, Inbred C57BL , Milk/adverse effects , Milk Hypersensitivity/blood , Milk Hypersensitivity/pathology
5.
Methods Mol Biol ; 2223: 337-355, 2021.
Article in English | MEDLINE | ID: mdl-33226603

ABSTRACT

Food allergy has been rising in prevalence over the last two decades, affecting more than 10% of the world population. Current management of IgE-mediated food allergy relies on avoidance and rescue medications; research into treatments that are safer and providing guaranteed and durable curative effects is, therefore, essential. T-cell epitope-based immunotherapy holds the potential for modulating food allergic responses without IgE cross-linking. In this chapter, we describe the methods in evaluating the therapeutic capacities of immunodominant T-cell epitopes in animal models of food allergy. Moreover, we explain in detail the methods to measure the allergen-specific antibody levels, prepare single-cell suspension from spleen, and prepare small intestine for immunohistochemical analysis of eosinophils and Foxp3+ cells.


Subject(s)
Allergens/administration & dosage , Desensitization, Immunologic/methods , Disease Models, Animal , Egg Hypersensitivity/therapy , Milk Hypersensitivity/therapy , Peptides/pharmacology , Shellfish Hypersensitivity/therapy , Adjuvants, Immunologic/administration & dosage , Administration, Oral , Aluminum Hydroxide/administration & dosage , Animals , Cholera Toxin/administration & dosage , Egg Hypersensitivity/immunology , Egg Hypersensitivity/pathology , Enzyme-Linked Immunosorbent Assay/methods , Eosinophils/drug effects , Eosinophils/immunology , Epitopes, T-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/immunology , Female , Humans , Immunoglobulin E/genetics , Immunoglobulin E/immunology , Immunohistochemistry/methods , Intestines/drug effects , Intestines/immunology , Mice, Inbred BALB C , Milk Hypersensitivity/immunology , Milk Hypersensitivity/pathology , Peptides/immunology , Shellfish Hypersensitivity/immunology , Shellfish Hypersensitivity/pathology , Spleen/drug effects , Spleen/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology
6.
Exp Neurol ; 334: 113460, 2020 12.
Article in English | MEDLINE | ID: mdl-32916172

ABSTRACT

Selective elimination of respiratory motor neurons using intrapleural injections of cholera toxin B fragment conjugated to saporin (CTB-SAP) mimics motor neuron death and respiratory deficits observed in rat models of neuromuscular diseases. This CTB-SAP model allows us to study the impact of motor neuron death on the output of surviving phrenic motor neurons. After 7(d) days of CTB-SAP, phrenic long-term facilitation (pLTF, a form of respiratory plasticity) is enhanced, but returns towards control levels at 28d. However, the mechanism responsible for this difference in magnitude of pLTF is unknown. In naïve rats, pLTF predominately requires 5-HT2 receptors, the new synthesis of BDNF, and MEK/ERK signaling; however, pLTF can alternatively be induced via A2A receptors, the new synthesis of TrkB, and PI3K/Akt signaling. Since A2A receptor-dependent pLTF is enhanced in naïve rats, we suggest that 7d CTB-SAP treated rats utilize the alternative mechanism for pLTF. Here, we tested the hypothesis that pLTF following CTB-SAP is: 1) TrkB and PI3K/Akt, not BDNF and MEK/ERK, dependent at 7d; and 2) BDNF and MEK/ERK, not TrkB and PI3K/Akt, dependent at 28d. Adult Sprague Dawley male rats were anesthetized, paralyzed, ventilated, and were exposed to acute intermittent hypoxia (AIH; 3, 5 min bouts of 10.5% O2) following bilateral, intrapleural injections at 7d and 28d of: 1) CTB-SAP (25 µg), or 2) un-conjugated CTB and SAP (control). Intrathecal C4 delivery included either: 1) small interfering RNA that targeted BDNF or TrkB mRNA; 2) UO126 (MEK/ERK inhibitor); or 3) PI828 (PI3K/Akt inhibitor). Our data suggest that pLTF in 7d CTB-SAP treated rats is elicited primarily through TrkB and PI3K/Akt-dependent mechanisms, whereas BDNF and MEK/ERK-dependent mechanisms induce pLTF in 28d CTB-SAP treated rats. This project increases our understanding of respiratory plasticity and its implications for breathing following motor neuron death.


Subject(s)
Cholera Toxin/toxicity , Long-Term Potentiation/physiology , Motor Neurons/physiology , Phrenic Nerve/physiology , Pleural Cavity/physiology , Saporins/toxicity , Animals , Cholera Toxin/administration & dosage , Long-Term Potentiation/drug effects , Male , Motor Neurons/drug effects , Motor Neurons/pathology , Phrenic Nerve/drug effects , Phrenic Nerve/pathology , Pleural Cavity/drug effects , Pleural Cavity/innervation , Rats , Rats, Sprague-Dawley , Saporins/administration & dosage
7.
Immunol Cell Biol ; 98(8): 667-681, 2020 09.
Article in English | MEDLINE | ID: mdl-32479679

ABSTRACT

Understanding the migration of lymphocytes to nonintestinal mucosal sites is fundamental to developing mucosal vaccination strategies. Studies have shown that nasal and oral immunization with cholera toxin (CT) stimulates, in addition to α4ß7+ , the induction of αE (CD103)ß7+ B cells. To determine the extent to which αE-associated ß7 contributes to antigen (Ag)-specific immunoglobulin (Ig)A responses in the upper respiratory tract, nasal CT vaccination was performed in wild-type (wt) and ß7-/- mice. At 16 days postprimary immunization, upper respiratory tract IgA responses were greater in ß7-/- mice than in wt mice. IgA induction by distal ß7-/- Peyer's patches, mesenteric lymph nodes and small intestinal lamina propria was minimal, in contrast to elevated gut IgA responses in wt mice. By 42 days postprimary immunization, ß7-/- gut IgA responses were restored, and upper respiratory tract Ag-specific IgA responses were equivalent to those of wt mice. Examination of homing receptor expression and cell-sorting experiments revealed that ß7-/- mice have increased usage of ß1 and αE integrins by upper respiratory tract B cells, suggesting that alternative integrins can facilitate lymphocyte migration to the upper respiratory tract, especially in the absence of ß7.


Subject(s)
B-Lymphocytes/immunology , Immunity, Mucosal , Immunoglobulin A , Integrin beta Chains , Administration, Intranasal , Animals , Cholera Toxin/administration & dosage , Integrin beta Chains/genetics , Intestinal Mucosa/immunology , Lymph Nodes/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Peyer's Patches/immunology , Vaccination/methods
8.
Chem Commun (Camb) ; 56(45): 6098-6101, 2020 Jun 07.
Article in English | MEDLINE | ID: mdl-32355935

ABSTRACT

Tracing of neurons plays an essential role in elucidating neural networks in the brain and spinal cord. Cholera toxin B subunit (CTB) is already widely used as a tracer although its use is limited by the need for immunohistochemical detection. A new construct incorporating non-canonical azido amino acids (azido-CTB) offers a novel way to expand the range and flexibility of this neuronal tracer. Azido-CTB can be detected rapidly in vivo following intramuscular tongue injection by 'click' chemistry, eliminating the need for antibodies. Cadmium selenide/zinc sulfide (CdSe/ZnS) core/shell nanoparticles were attached to azido-CTB by strain-promoted alkyne-azide cycloaddition to make a nano-conjugate. Following tongue injections the complex was detected in vivo in the brainstem by light microscopy and electron microscopy via silver enhancement. This method does not require membrane permeabilization and so ultrastructure is maintained. Azido-CTB offers new possibilities to enhance the utility of CTB as a neuronal tracer and delivery vehicle by modification using 'click' chemistry.


Subject(s)
Azides/administration & dosage , Cadmium Compounds/administration & dosage , Cholera Toxin/administration & dosage , Motor Neurons/metabolism , Nanoparticles/administration & dosage , Selenium Compounds/administration & dosage , Sulfides/administration & dosage , Zinc Compounds/administration & dosage , Animals , Azides/chemistry , Brain Stem/metabolism , Cadmium Compounds/chemistry , Cholera Toxin/chemistry , Mice , Nanoparticles/chemistry , Selenium Compounds/chemistry , Sulfides/chemistry , Zinc Compounds/chemistry
9.
Int J Pharm ; 582: 119348, 2020 May 30.
Article in English | MEDLINE | ID: mdl-32325240

ABSTRACT

Influenza vaccines administered intramuscularly exhibit poor mucosal immune responses in the respiratory tract which is the prime site of the infection. Intranasal vaccination is a potential route for vaccine delivery which has been demonstrated effective in inducing protective immune responses in both systemic and mucosal compartments. For this purpose, nanoparticles have been used as antigen delivery systems to improve antigen capture by immune cells. In this paper we demonstrate efficient delivery of viral antigens to airway epithelial cells, macrophages and dendritic cells, using polysaccharide nanoparticles (NPL), leading to a strong protection against influenza virus infection. A formulation combining split Udorn virus antigens with NPL and the mucosal protein adjuvant CTA1-DD was administered intranasally and resulted in an enhanced specific humoral immune response. Furthermore, NPL carrying split Udorn, with or without CTA1-DD, inhibited virus transmission from infected to uninfected naive mice. These results demonstrate that an intranasal delivery system combining NPL, mucosal adjuvant CTA1-DD and split virus antigens confers robust protection against influenza infection and inhibits virus transmission.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Antigens, Viral/administration & dosage , Cholera Toxin/administration & dosage , Drug Carriers , Influenza A Virus, H3N2 Subtype/immunology , Influenza Vaccines/administration & dosage , Nanoparticles , Orthomyxoviridae Infections/prevention & control , Polysaccharides/chemistry , Recombinant Fusion Proteins/administration & dosage , Adjuvants, Immunologic/chemistry , Administration, Intranasal , Animals , Antibodies, Viral/blood , Antigens, Viral/chemistry , Antigens, Viral/immunology , Cholera Toxin/chemistry , Cholera Toxin/immunology , Disease Models, Animal , Drug Compounding , Immunity, Humoral/drug effects , Influenza Vaccines/chemistry , Influenza Vaccines/immunology , Mice, Inbred BALB C , Mice, Inbred DBA , Orthomyxoviridae Infections/transmission , Orthomyxoviridae Infections/virology , Porosity , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/immunology
10.
Sci Rep ; 10(1): 893, 2020 01 21.
Article in English | MEDLINE | ID: mdl-31964988

ABSTRACT

Intramuscular injections of botulinum toxin block pre-synaptic cholinergic release at neuromuscular junctions producing a temporary paralysis of affected motor units. There is increasing evidence, however, that the effects are not restricted to the periphery and can alter the central excitability of the motoneurones at the spinal level. This includes increases in input resistance, decreases in rheobase currents for action potentials and prolongations of the post-spike after-hyperpolarization. The aim of our experiments was to investigate possible anatomical explanations for these changes. Unilateral injections of Botulinum toxin A mixed with a tracer were made into the gastrocnemius muscle of adult rats and contralateral tracer only injections provided controls. Immunohistochemistry for Ankyrin G and the vesicular acetylcholine transporter labelled axon initial segments and cholinergic C-boutons on traced motoneurones at 2 weeks post-injection. Soma size was not affected by the toxin; however, axon initial segments were 5.1% longer and 13.6% further from the soma which could explain reductions in rheobase. Finally, there was a reduction in surface area (18.6%) and volume (12.8%) but not frequency of C-boutons on treated motoneurones potentially explaining prolongations of the after-hyperpolarization. Botulinum Toxin A therefore affects central anatomical structures controlling or modulating motoneurone excitability explaining previously observed excitability changes.


Subject(s)
Axon Initial Segment/drug effects , Botulinum Toxins, Type A/pharmacology , Motor Neurons/drug effects , Muscle, Skeletal/drug effects , Animals , Botulinum Toxins, Type A/administration & dosage , Cholera Toxin/administration & dosage , Cholinergic Neurons/drug effects , Injections, Intramuscular , Male , Motor Neurons/physiology , Muscle, Skeletal/cytology , Rats, Wistar , Spinal Cord/cytology , Vesicular Acetylcholine Transport Proteins/metabolism
11.
Mucosal Immunol ; 12(6): 1268-1279, 2019 11.
Article in English | MEDLINE | ID: mdl-31501516

ABSTRACT

Our understanding of how class-switch recombination (CSR) to IgA occurs in the gut is still incomplete. Earlier studies have indicated that Tregs are important for IgA CSR and these cells were thought to transform into follicular helper T cells (Tfh), responsible for germinal center formation in the Peyer's patches (PP). Following adoptive transfer of T-cell receptor-transgenic (TCR-Tg) CD4 T cells into nude mice, we unexpectedly found that oral immunization did not require an adjuvant to induce strong gut IgA and systemic IgG responses, suggesting an altered regulatory environment in the PP. After sorting of splenic TCR-Tg CD4 T cells into CD25+ or CD25- cells we observed that none of these fractions supported a gut IgA response, while IgG responses were unperturbed in mice receiving the CD25- cell fraction. Hence, while Tfh functions resided in the CD25- fraction the IgA CSR function in the PP was dependent on CD25+ Foxp3+ Tregs, which were found to be Helios+ neuropilin-1+ thymus-derived Tregs. This is the first study to demonstrate that Tfh and IgA CSR functions are indeed, unique, and separate functions in the PP with the former being TCR-dependent while the latter appeared to be antigen independent.


Subject(s)
B-Lymphocytes/immunology , Immunoglobulin A/genetics , Immunoglobulin Class Switching , Peyer's Patches/immunology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Regulatory/immunology , Thymus Gland/immunology , Adjuvants, Immunologic/administration & dosage , Adoptive Transfer , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , Cells, Cultured , Cholera Toxin/administration & dosage , Coculture Techniques , DNA-Binding Proteins/immunology , DNA-Binding Proteins/metabolism , Immunization , Immunoglobulin A/immunology , Immunoglobulin A/metabolism , Immunoglobulin Class Switching/drug effects , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Mice, SCID , Mice, Transgenic , Neuropilin-1/immunology , Neuropilin-1/metabolism , Ovalbumin/administration & dosage , Peyer's Patches/metabolism , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Helper-Inducer/transplantation , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/transplantation , Thymus Gland/drug effects , Thymus Gland/metabolism , Transcription Factors/immunology , Transcription Factors/metabolism
12.
Methods Mol Biol ; 2021: 201-215, 2019.
Article in English | MEDLINE | ID: mdl-31309507

ABSTRACT

Proteus mirabilis is a major cause of complicated urinary tract infections (UTIs). P. mirabilis' urease activity hydrolyzes urea and raises urine pH levels, which can catalyze bladder and kidney stone formation. This urolithiasis leads to harder-to-treat infections, possible urinary blockage, and subsequent septicemia. Development of a mucosal vaccine against P. mirabilis urinary tract infections is critical to protect against this potentially deadly infection process. Here, we describe the methodology necessary to produce a vaccine candidate conjugated to cholera toxin, administer the vaccine via the intranasal route, and test efficacy in a murine transurethral P. mirabilis infection model.


Subject(s)
Antibodies, Bacterial/metabolism , Immunotoxins/administration & dosage , Proteus Infections/prevention & control , Proteus mirabilis/immunology , Urinary Tract Infections/prevention & control , Administration, Intranasal , Animals , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/immunology , Cholera Toxin/administration & dosage , Cholera Toxin/immunology , Disease Models, Animal , Female , Humans , Immunotoxins/immunology , Mice , Mice, Inbred CBA , Vaccination
13.
J Comput Biol ; 26(10): 1177-1190, 2019 10.
Article in English | MEDLINE | ID: mdl-31120321

ABSTRACT

Helicobacter pylori is an infectious agent that colonizes the gastric mucosa of half of the population worldwide. This bacterium has been recognized as belonging to group 1 carcinogen by the World Health Organization for the role in development of gastritis, peptic ulcers, and cancer. Due to the increase in resistance to antibiotics used in the anti-H. pylori therapy, the development of an effective vaccine is an alternative of great interest, which remains a challenge. Therefore, a rational, strategic, and efficient vaccine design against H. pylori is necessary where the use of the most current bioinformatics tools could help achieve it. In this study, immunoinformatics approach was used to design a novel multiepitope oral vaccine against H. pylori. Our multiepitope vaccine is composed of cholera toxin subunit B (CTB) that is used as a mucosal adjuvant to enhance vaccine immunogenicity for oral immunization. CTB fused to 11 epitopes predicted of pathogenic (UreB170-189, VacA459-478, CagA1103-1122, GGT106-126, NapA30-44, and OipA211-230) and colonization (HpaA33-52, FlaA487-506, FecA437-456, BabA129-149, and SabA540-559) proteins from H. pylori. CKS9 peptide (CKSTHPLSC) targets epithelial microfold cells to enhance vaccine uptake from the gut barrier. All sequences were joined to each other by proper linkers. The vaccine was modeled and validated to achieve a high-quality three-dimensional structure. The vaccine design was evaluated as nonallergenic, antigenic, soluble, and with an appropriate molecular weight and isoelectric point. Our results suggest that our newly designed vaccine could serve as a promising anti-H. pylori vaccine candidate.


Subject(s)
Bacterial Proteins/immunology , Bacterial Vaccines/immunology , Epitopes/immunology , Helicobacter Infections/prevention & control , Helicobacter pylori/immunology , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/pharmacology , Administration, Oral , Amino Acid Sequence , Animals , Bacterial Proteins/administration & dosage , Bacterial Proteins/chemistry , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/chemistry , Cholera Toxin/administration & dosage , Cholera Toxin/immunology , Computational Biology , Epitopes/administration & dosage , Helicobacter Infections/immunology , Helicobacter pylori/chemistry , Humans , Mice , Models, Molecular , Protein Structure, Secondary
14.
J Biotechnol ; 296: 83-92, 2019 Apr 20.
Article in English | MEDLINE | ID: mdl-30898688

ABSTRACT

Silkworms are an economically important insect.Silkworm pupae are also a nutrient-rich food and can be used as a pharmaceutical intermediate.The N-terminus of Aß includes 1-15 amino acid residues with a B cell surface antigen that is necessary to produce antibody and prevent the adverse reactions observed in response to the full Aß42 peptide. In this study, we used silkworm pupae to develop a safer vaccine for Alzheimer's disease (AD) patients. Aß15 peptide was fused with the cholera toxin B subunit (CTB) and expressed in silkworm pupae. Then, we tested an oral vaccine with the peptide expressed by silkworm pupae in a transgenic mouse model of AD. The results show that anti-Aß antibodies were induced, Aß deposition in the brain decreased, the content of malondialdehyde was lower than in the other group, and memory and cognition of the mice improved. These results suggest that the high-nutrient CTB-Aß15 silkworm pupa vaccine has a potential clinical application for the prevention of AD.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/genetics , Cholera Toxin/administration & dosage , Vaccines/administration & dosage , Alzheimer Disease/genetics , Alzheimer Disease/immunology , Alzheimer Disease/pathology , Amyloid beta-Peptides/immunology , Animals , Bombyx/chemistry , Bombyx/genetics , Cholera Toxin/genetics , Cholera Toxin/immunology , Disease Models, Animal , Gene Expression Regulation/genetics , Humans , Mice , Mice, Transgenic/genetics , Protein Isoforms/chemistry , Protein Isoforms/genetics , Pupa/genetics , Pupa/growth & development , Vaccines/genetics , Vaccines/immunology
15.
Med Microbiol Immunol ; 208(1): 89-100, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30209565

ABSTRACT

Escherichia coli O157:H7 with its traits such as intestinal colonization and fecal-oral route of transmission demands mucosal vaccine development. E. coli secreted protein B (EspB) is one of the key type III secretory system (TTSS) targets for mucosal candidate vaccine due to its indispensable role in the pathogenesis of E. coli O157:H7. However, mucosally administered recombinant proteins have low immunogenicity which could be overcome by the use of mucosal adjuvants. The quest for safe, potent mucosal adjuvant has recognized ΔG fragment of Zonula occludens toxin of Vibrio cholerae with such properties. ΔG enhances mucosal permeability via the paracellular route by altering epithelial tight junction structure in a reversible, ephemeral and non-toxic manner. Therefore, we tested whether recombinant ΔG intranasally co-administered with truncated EspB (EspB + ΔG) could serve as an effective mucosal adjuvant. Results showed that EspB + ΔG group induced higher systemic IgG and mucosal IgA than EspB alone. Moreover, EspB alone developed Th2 type response with IgG1/IgG2a ratio (1.64) and IL-4, IL-10 cytokines whereas that of EspB + ΔG group generated mixed Th1/Th2 type immune response evident from IgG1/IgG2a ratio (1.17) as well as IL-4, IL-10 and IFN-γ cytokine levels compared to control. Sera of EspB + ΔG group inhibited TTSS mediated haemolysis of murine RBCs more effectively compared to EspB, control group and sera of both EspB + ΔG, EspB group resulted in similar levels of efficacious reduction in E. coli O157:H7 adherence to Caco-2 cells compared to control. Moreover, vaccination with EspB + ΔG resulted in significant reduction in E. coli O157:H7 fecal shedding compared to EspB and control group in experimentally challenged streptomycin-treated mice. These results demonstrate mucosal adjuvanticity of ΔG co-administered with EspB in enhancing overall immunogenicity to reduce E. coli O157:H7 shedding.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Bacterial Outer Membrane Proteins/immunology , Cholera Toxin/administration & dosage , Escherichia coli Infections/prevention & control , Escherichia coli O157/immunology , Escherichia coli Proteins/immunology , Immunity, Humoral , Immunity, Mucosal , Administration, Intranasal , Animals , Antibodies, Bacterial/analysis , Antibodies, Bacterial/blood , Bacterial Adhesion/drug effects , Bacterial Outer Membrane Proteins/administration & dosage , Bacterial Shedding , Caco-2 Cells , Cholera Toxin/genetics , Disease Transmission, Infectious , Endotoxins , Escherichia coli Infections/immunology , Escherichia coli Infections/microbiology , Escherichia coli Proteins/administration & dosage , Escherichia coli Vaccines/administration & dosage , Escherichia coli Vaccines/immunology , Feces/microbiology , Humans , Immunoglobulin A/analysis , Immunoglobulin G/blood , Mice, Inbred BALB C , Mutant Proteins/administration & dosage , Mutant Proteins/genetics , Recombinant Proteins/administration & dosage , Recombinant Proteins/immunology , Th2 Cells/immunology
16.
J Allergy Clin Immunol ; 143(3): 1027-1037.e7, 2019 03.
Article in English | MEDLINE | ID: mdl-30342892

ABSTRACT

BACKGROUND: Improved animal models are needed to understand the genetic and environmental factors that contribute to food allergy. OBJECTIVE: We sought to assess food allergy phenotypes in a genetically diverse collection of mice. METHODS: We selected 16 Collaborative Cross (CC) mouse strains, as well as the classic inbred C57BL/6J, C3H/HeJ, and BALB/cJ strains, for screening. Female mice were sensitized to peanut intragastrically with or without cholera toxin and then challenged with peanut by means of oral gavage or intraperitoneal injection and assessed for anaphylaxis. Peanut-specific immunoglobulins, T-cell cytokines, regulatory T cells, mast cells, and basophils were quantified. RESULTS: Eleven of the 16 CC strains had allergic reactions to intraperitoneal peanut challenge, whereas only CC027/GeniUnc mice reproducibly experienced severe symptoms after oral food challenge (OFC). CC027/GeniUnc, C3H/HeJ, and C57BL/6J mice all mounted a TH2 response against peanut, leading to production of IL-4 and IgE, but only the CC027/GeniUnc mice reacted to OFC. Orally induced anaphylaxis in CC027/GeniUnc mice was correlated with serum levels of Ara h 2 in circulation but not with allergen-specific IgE or mucosal mast cell protease 1 levels, indicating systemic allergen absorption is important for anaphylaxis through the gastrointestinal tract. Furthermore, CC027/GeniUnc, but not C3H/HeJ or BALB/cJ, mice can be sensitized in the absence of cholera toxin and react on OFC to peanut. CONCLUSIONS: We have identified and characterized CC027/GeniUnc mice as a strain that is genetically susceptible to peanut allergy and prone to severe reactions after OFC. More broadly, these findings demonstrate the untapped potential of the CC population in developing novel models for allergy research.


Subject(s)
Allergens/adverse effects , Arachis/adverse effects , Peanut Hypersensitivity/genetics , Allergens/immunology , Animals , Arachis/immunology , Cholera Toxin/administration & dosage , Cytokines/genetics , Female , Genetic Variation , Immunoglobulin E/immunology , Jejunum/immunology , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Peanut Hypersensitivity/immunology , Species Specificity , Spleen/immunology
17.
Front Immunol ; 9: 2212, 2018.
Article in English | MEDLINE | ID: mdl-30319653

ABSTRACT

CD4+ T cells are major players in the immune response against several diseases; including AIDS, leishmaniasis, tuberculosis, influenza and cancer. Their activation has been successfully achieved by administering antigen coupled with antibodies, against DC-specific receptors in combination with adjuvants. Unfortunately, most of the adjuvants used so far in experimental models are unsuitable for human use. Therefore, human DC-targeted vaccination awaits the description of potent, yet nontoxic adjuvants. The nontoxic cholera B subunit (CTB) can be safely used in humans and it has the potential to activate CD4+ T cell responses. However, it remains unclear whether CTB can promote DC activation and can act as an adjuvant for DC-targeted antigens. Here, we evaluated the CTB's capacity to activate DCs and CD4+ T cell responses, and to generate long-lasting protective immunity. Intradermal (i.d.) administration of CTB promoted late and prolonged activation and accumulation of skin and lymphoid-resident DCs. When CTB was co-administered with anti-DEC205-OVA, it promoted CD4+ T cell expansion, differentiation, and infiltration to peripheral nonlymphoid tissues, i.e., the skin, lungs and intestine. Indeed, CTB promoted a polyfunctional CD4+ T cell response, including the priming of Th1 and Th17 cells, as well as resident memory T (RM) cell differentiation in peripheral nonlymphoid tissues. It is worth noting that CTB together with a DC-targeted antigen promoted local and systemic protection against experimental melanoma and murine rotavirus. We conclude that CTB administered i.d. can be used as an adjuvant to DC-targeted antigens for the induction of broad CD4+ T cell responses as well as for promoting long-lasting protective immunity.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Cholera Toxin/administration & dosage , Dendritic Cells/immunology , Lectins, C-Type/antagonists & inhibitors , Receptors, Cell Surface/antagonists & inhibitors , Vaccination/methods , Animals , Antigens, CD/immunology , Cell Line, Tumor/transplantation , Disease Models, Animal , Female , Humans , Injections, Intradermal , Lectins, C-Type/immunology , Lymphocyte Activation/immunology , Male , Melanoma/immunology , Melanoma/prevention & control , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Minor Histocompatibility Antigens/immunology , Receptors, Cell Surface/immunology , Rotavirus/immunology , Rotavirus Infections/immunology , Rotavirus Infections/prevention & control , Rotavirus Infections/virology , Th1 Cells/immunology , Th17 Cells/immunology , Treatment Outcome
18.
Neuroscience ; 390: 303-316, 2018 10 15.
Article in English | MEDLINE | ID: mdl-30179644

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a devastating disease leading to degeneration of motor neurons and skeletal muscles, including those required for swallowing. Tongue weakness is one of the earliest signs of bulbar dysfunction in ALS, which is attributed to degeneration of motor neurons in the hypoglossal nucleus in the brainstem, the axons of which directly innervate the tongue. Despite its fundamental importance, dysphagia (difficulty swallowing) and strategies to preserve swallowing function have seldom been studied in ALS models. It is difficult to study dysphagia in ALS models since the amount and rate at which hypoglossal motor neuron death occurs cannot be controlled, and degeneration is not limited to the hypoglossal nucleus. Here, we report a novel experimental model using intralingual injections of cholera toxin B conjugated to saporin (CTB-SAP) to study the impact of only hypoglossal motor neuron death without the many complications that are present in ALS models. Hypoglossal motor neuron survival, swallowing function, and hypoglossal motor output were assessed in Sprague-Dawley rats after intralingual injection of either CTB-SAP (25 g) or unconjugated CTB and SAP (controls) into the genioglossus muscle. CTB-SAP treated rats exhibited significant (p ≤ 0.05) deficits vs. controls in: (1) lick rate (6.0 ±â€¯0.1 vs. 6.6 ±â€¯0.1 Hz; (2) hypoglossal motor output (0.3 ±â€¯0.05 vs. 0.6 ±â€¯0.10 mV); and (3) hypoglossal motor neuron survival (398 ±â€¯34 vs. 1018 ±â€¯41 neurons). Thus, this novel, inducible model of hypoglossal motor neuron death mimics the dysphagia phenotype that is observed in ALS rodent models, and will allow us to study strategies to preserve swallowing function.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , Cholera Toxin/administration & dosage , Deglutition Disorders/pathology , Disease Models, Animal , Hypoglossal Nerve/pathology , Motor Neurons/pathology , Saporins/administration & dosage , Animals , Cell Death , Deglutition Disorders/chemically induced , Hypoglossal Nerve/drug effects , Hypoglossal Nerve/physiopathology , Male , Motor Neurons/drug effects , Motor Neurons/physiology , Rats, Sprague-Dawley , Tongue/drug effects , Tongue/innervation
19.
Microb Pathog ; 124: 170-177, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30138759

ABSTRACT

Vibrio cholera is a Gram-negative pathogen that causes diarrheal disease. The B subunit of Chlora toxin (CtxB) is one of the most important antigens of Vibrio cholera in which mediates the attachment of the bacteria to target cells. The aim of this study was to prepare chitosan nanoparticles containing CtxB and evaluate the effect of the antigen entrapment on the immunogenicity of this antigen. For this, the pET28a vector was induced using IPTG. Recombinant CtxB protein was expressed and purified using Ni-NTA column and finally was confirmed by western blotting. Following the confirmation of the protein entrapment onto the chitosan nanoparticles, the formulation was prescribed to BALB/c mice in three groups, including oral, oral-injection and injection groups. Serum and fecal IgA and IgG were evaluated by ELISA test. Finally, challenge of immunized mice was performed using Ctx toxin and rabbit ileal loop test. Using SDS-PAGE and western blotting, the 17.5 kDa recombinant CtxB was confirmed. Size electrical charge and of nanoparticles were determined and approved by Zetasizer. Nanoparticles prescription showed 1/102400 IgG endpoint titers for injection groups and 1/1600, 1/6400 for oral, oral-injection groups respectively and Serum and fecal IgA endpoint titers showed above 1/160 in all groups. Furthermore, immunized mice were able to neutralize Ctx toxin by ileal loop test. The CtxB is a suitable immunogen of V. cholera to be incorporated in both protective and preventive vaccines. Chitosan nanoparticles improve the immune responses and it may be used as a carrier for vaccine delivery.


Subject(s)
Antigens, Bacterial/immunology , Cholera Toxin/immunology , Cholera/prevention & control , Nanoparticles/chemistry , Vibrio cholerae/immunology , Animals , Antibodies, Bacterial/immunology , Antigens, Bacterial/administration & dosage , Antigens, Bacterial/genetics , Chitosan/administration & dosage , Chitosan/chemistry , Cholera/immunology , Cholera/microbiology , Cholera Toxin/administration & dosage , Cholera Toxin/chemistry , Drug Evaluation, Preclinical , Female , Humans , Mice , Mice, Inbred BALB C , Nanoparticles/administration & dosage , Rabbits , Vibrio cholerae/chemistry , Vibrio cholerae/genetics
20.
Int Immunopharmacol ; 63: 211-219, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30103196

ABSTRACT

Ruminants are the major reservoirs of Escherichia coli O157:H7 and its fecal shedding mainly act as a source of entry of this pathogen into the human food chain. In humans, E. coli O157:H7 infection causes diarrhea, hemorrhagic colitis and hemolytic uremic syndrome. Intimate adherence of E. coli O157:H7 is mediated by Translocated intimin receptor (Tir) to which intimin binds in the host cell. Since E. coli O157:H7 colonizes intestinal epithelium, the mucosal vaccine has a potential to prevent its colonization. Zonula occludens toxin (Zot) of Vibrio cholerae transiently, reversibly alters epithelial tight junction structure to increase mucosal permeability of macromolecules via paracellular route. The C-terminal region of Zot (ΔG) responsible for this function could be used for mucosal antigen delivery. Therefore, we employed individual (Tir), cocktail (ΔG + Tir), fusion protein (ΔG-Tir) and assessed the efficacy of its intranasal immunization on immunogenicity and fecal shedding of E. coli O157:H7 in streptomycin treated mouse model. Compared to control, ΔG + Tir, ΔG-Tir immunized mice elicited significant antigen specific antibody titers in serum (IgG, IgA) and feces (IgA), whereas Tir immunized mice induced only serum IgG titer. Cytokine analysis revealed mixed Th1/Th2 type immune response in case of ΔG + Tir, ΔG-Tir group while that of Tir group was solely Th2 type. Tir, ΔG + Tir and ΔG-Tir immunized mice showed reduction in shedding of E. coli O157:H7 compared to control group. However, ΔG-Tir immunized group performed better than ΔG + Tir, Tir group in reducing fecal shedding. Overall, our results demonstrate that intranasal immunization of ΔG-Tir induces effective systemic, mucosal, cellular immune responses and represents a promising mucosal subunit vaccine to prevent E. coli O157:H7 colonization.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Cholera Toxin/administration & dosage , Escherichia coli Proteins/administration & dosage , Receptors, Cell Surface/administration & dosage , Recombinant Fusion Proteins/administration & dosage , Vaccines, Subunit/administration & dosage , Administration, Intranasal , Animals , Antibodies, Bacterial/blood , Cytokines/immunology , Endotoxins , Escherichia coli Infections/immunology , Escherichia coli O157 , Feces/microbiology , Female , Immunization , Immunoglobulin A/blood , Immunoglobulin G/blood , Mice, Inbred BALB C
SELECTION OF CITATIONS
SEARCH DETAIL
...