Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 90
Filter
1.
Toxins (Basel) ; 13(12)2021 11 29.
Article in English | MEDLINE | ID: mdl-34941688

ABSTRACT

The social wasp Polybia paulista (Hymenoptera, Vespidae) is highly aggressive, being responsible for many medical occurrences. One of the most allergenic components of this venom is Antigen 5 (Poly p 5). The possible modulation of the in vitro immune response induced by antigen 5 from P. paulista venom, expressed recombinantly (rPoly p 5), on BALB/c mice peritoneal macrophages, activated or not with LPS, was assessed. Here, we analyzed cell viability changes, expression of the phosphorylated form of p65 NF-κB subunit, nitric oxide (NO), proinflammatory cytokines production, and co-stimulatory molecules (CD80, CD86). The results suggest that rPoly p 5 does not affect NO production nor the expression of co-stimulatory molecules in mouse peritoneal macrophages. On the other hand, rPoly p 5 induced an increase in IL-1ß production in non-activated macrophages and a reduction in the production of TNF-α and MCP-1 cytokines in activated macrophages. rPoly p 5 decreased the in vitro production of the phosphorylated p65 NF-κB subunit in non-activated macrophages. These findings suggest an essential role of this allergen in the polarization of functional M2 macrophage phenotypes, when analyzed in previously activated macrophages. Further investigations, mainly in in vivo studies, should be conducted to elucidate Polybia paulista Ag5 biological role in the macrophage functional profile modulation.


Subject(s)
Antigens/toxicity , Macrophages, Peritoneal/drug effects , Wasp Venoms/chemistry , Wasps/physiology , Animals , Gene Expression Regulation/drug effects , Mice , Mice, Inbred BALB C , Nitric Oxide , Phosphorylation , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism , Wasp Venoms/toxicity
2.
Nanotoxicology ; 14(5): 711-724, 2020 06.
Article in English | MEDLINE | ID: mdl-32374645

ABSTRACT

Prenatal particle exposure has been shown to increase allergic responses in offspring. Carbon nanotubes (CNTs) possess immunomodulatory properties, but it is unknown whether maternal exposure to CNTs interferes with offspring immune development. Here, C57Bl/6J female mice were intratracheally instilled with 67 of µg multiwalled CNTs on the day prior to mating. After weaning, tolerance and allergy responses were assessed in the offspring. Offspring of CNT-exposed (CNT offspring) and of sham-exposed dams (CTRL offspring) were intranasally exposed to ovalbumin (OVA) once weekly for 5 weeks to induce airway mucosal tolerance. Subsequent OVA sensitization and aerosol inhalation caused low or no OVA-specific IgE production and no inflammation. However, the CNT offspring presented with significantly lower OVA-specific IgG1 levels than CTRL offspring. In other groups of 5-week-old offspring, low-dose sensitization with OVA and subsequent OVA aerosol inhalation led to significantly lower OVA-specific IgG1 production in CNT compared to CTRL offspring. OVA-specific IgE and airway inflammation were non-significantly reduced in CNT offspring. The immunomodulatory effects of pre-gestational exposure to multiwalled CNTs were unexpected, but very consistent. The observations of suppressed antigen-specific IgG1 production may be of importance for infection or vaccination responses and warrant further investigation.


Subject(s)
Antibody Formation/drug effects , Antigens/toxicity , Hypersensitivity/etiology , Nanotubes, Carbon/toxicity , Prenatal Exposure Delayed Effects/chemically induced , Animals , Antigens/chemistry , Female , Humans , Hypersensitivity/immunology , Immune Tolerance/drug effects , Immune Tolerance/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Inflammation , Maternal Exposure/adverse effects , Mice , Mice, Inbred BALB C , Nanotubes, Carbon/chemistry , Ovalbumin/immunology , Pregnancy , Prenatal Exposure Delayed Effects/immunology
3.
Toxins (Basel) ; 10(11)2018 Nov 12.
Article in English | MEDLINE | ID: mdl-30424519

ABSTRACT

Ricin, a highly lethal toxin derived from the seeds of Ricinus communis (castor beans) is considered a potential biological threat agent due to its high availability, ease of production, and to the lack of any approved medical countermeasure against ricin exposures. To date, the use of neutralizing antibodies is the most promising post-exposure treatment for ricin intoxication. The aim of this work was to generate anti-ricin antitoxin that confers high level post-exposure protection against ricin challenge. Due to safety issues regarding the usage of ricin holotoxin as an antigen, we generated an inactivated toxin that would reduce health risks for both the immunizer and the immunized animal. To this end, a monomerized ricin antigen was constructed by reducing highly purified ricin to its monomeric constituents. Preliminary immunizing experiments in rabbits indicated that this monomerized antigen is as effective as the native toxin in terms of neutralizing antibody elicitation and protection of mice against lethal ricin challenges. Characterization of the monomerized antigen demonstrated that the irreversibly detached A and B subunits retain catalytic and lectin activity, respectively, implying that the monomerization process did not significantly affect their overall structure. Toxicity studies revealed that the monomerized ricin displayed a 250-fold decreased activity in a cell culture-based functionality test, while clinical signs were undetectable in mice injected with this antigen. Immunization of a horse with the monomerized toxin was highly effective in elicitation of high titers of neutralizing antibodies. Due to the increased potential of IgG-derived adverse events, anti-ricin F(ab')2 antitoxin was produced. The F(ab')2-based antitoxin conferred high protection to intranasally ricin-intoxicated mice; ~60% and ~34% survival, when administered 24 and 48 h post exposure to a lethal dose, respectively. In line with the enhanced protection, anti-inflammatory and anti-edematous effects were measured in the antitoxin treated mice, in comparison to mice that were intoxicated but not treated. Accordingly, this anti-ricin preparation is an excellent candidate for post exposure treatment of ricin intoxications.


Subject(s)
Antigens/toxicity , Antitoxins/therapeutic use , Ricin/toxicity , Animals , Antibodies, Neutralizing/immunology , Antigens/immunology , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/immunology , Female , Horses , Mice , Rabbits , Ricin/immunology , Vaccination
4.
Mol Immunol ; 93: 1-8, 2018 01.
Article in English | MEDLINE | ID: mdl-29112834

ABSTRACT

This study examined whether NK cells are important for resolution of antigen-induced inflammation. C57BL/6 mice were immunized twice with methylated BSA (mBSA) and inflammation induced by intraperitoneal injection of mBSA. Mice were injected intravenously with anti-asialo GM1 (αASGM1) or a control antibody 24h prior to peritonitis induction and peritoneal exudate collected at different time points. Expression of surface molecules and apoptosis on peritoneal cells was determined by flow cytometry and concentration of chemokines, cytokines, soluble cytokine receptors and lipid mediators by ELISA and LC-MS/MS. Apoptosis in parathymic lymph nodes and spleens was determined by TUNEL staining. Mice administered αASGM1 had lower peritoneal NK cell numbers and a higher number of peritoneal neutrophils 12h after induction of inflammation than control mice. The number of neutrophils was still high in the αASGM1 treated mice when their number had returned to baseline levels in the control mice, 48h after induction of inflammation. Peritoneal concentrations of the neutrophil regulators G-CSF and IL-12p40 were higher at 12h in the αASGM1 treated mice than in the control mice, whereas concentrations of lipid mediators implicated in resolution of inflammation, i.e. LXA4 and PGE2, were lower. Reduced apoptosis was detected in peritoneal neutrophils as well as in draining lymph nodes and spleens from the αASGM1 treated mice compared with that in the control mice. In addition, αASGM1 treated mice had lower number of peritoneal NK cells expressing NKp46 and NKG2D, receptors implicated in NK cell-induced neutrophil apoptosis. Furthermore, αASGM1 treatment completely blocked the increase in CD27+ NK cells that occurred in control mice following induction of inflammation, but CD27+ NK cells have been suggested to have a regulatory role. These results indicate a crucial role for NK cells in resolution of antigen-induced inflammation and suggest their importance in tempering neutrophil recruitment and maintaining neutrophil apoptosis.


Subject(s)
Antigens/toxicity , Killer Cells, Natural/immunology , Peritonitis/immunology , Animals , Antibodies/immunology , Antibodies/therapeutic use , Apoptosis/drug effects , Chemokines/analysis , Dinoprostone/analysis , Female , G(M1) Ganglioside/antagonists & inhibitors , G(M1) Ganglioside/immunology , Granulocyte Colony-Stimulating Factor/analysis , Immunophenotyping , Inflammation Mediators/analysis , Interleukin-12 Subunit p40/analysis , Killer Cells, Natural/drug effects , Lipoxins/analysis , Lymph Nodes/pathology , Mice , Mice, Inbred C57BL , Neutrophil Infiltration/drug effects , Peritonitis/chemically induced , Peritonitis/metabolism , Peritonitis/therapy , Receptors, Natural Killer Cell/analysis , Serum Albumin, Bovine/toxicity , Spleen/pathology
5.
BMC Pulm Med ; 16: 50, 2016 Apr 14.
Article in English | MEDLINE | ID: mdl-27075111

ABSTRACT

BACKGROUND: This study aims to find out whether extracellular miRNAs is implicated in recurrent childhood wheezing with asthmatic risk. METHODS: One hundred and forty children of Chinese Han population were recruited for this study. Plasma and intracellular miRNAs from children with recurrent wheezing and rats with antigen induced pulmonary inflammation (AIPI) were detected by using reverse transcription-quantitative PCR. Differential leukocytes in blood were automatically counted. Total IgE was detected by enzyme-linked immunosorbent assay. Clinical implication in diagnosis was evaluated using receiver operating characteristic curves. RESULTS: The increase of plasma miR-21 and miR-26a was screened out from 11 candidate miRNAs and validated in wheezing children. The level of expression for both miRNAs were comparable in different age and gender. Plasma miR-21 was more preferable to miR-26a and total IgE for diagnosis. Plasma miR-21 and miR-26a levels were not significantly correlated with various leukocyte counts or miRNA expression in blood cells. In acute and chronic AIPI rats, miR-21 levels increased in both plasma and lavaged lung compared with control. Moreover, circulating miR-21 and miR-26a levels were highly positively correlated with infiltrated cell counts in bronchoalveolar lavage fluid of AIPI rats. CONCLUSIONS: Circulating miR-21 and miR-26a increase in wheezing children and AIPI rats. This not only manifests their strong clinical implication in recurrent childhood wheezing with asthma risk, but also provides novel insights into the role of extracellular miRNAs during development of airway inflammation and recurrent wheezing.


Subject(s)
MicroRNAs/genetics , Pneumonia/genetics , Respiratory Sounds/genetics , Animals , Antigens/immunology , Antigens/toxicity , Asian People , Child , Child, Preschool , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Forced Expiratory Volume , Humans , Immunoglobulin E/immunology , Infant , Male , MicroRNAs/blood , Ovalbumin/immunology , Ovalbumin/toxicity , Pneumonia/chemically induced , Pneumonia/immunology , Rats , Recurrence , Respiratory Sounds/immunology , Respiratory Sounds/physiopathology , Reverse Transcriptase Polymerase Chain Reaction , Vital Capacity
6.
Endocrinology ; 157(1): 16-22, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26517045

ABSTRACT

Activating autoantibodies to the ß1-adrenergic and M2 muscarinic receptors are present in a very high percentage of patients with Graves' disease and atrial fibrillation (AF). The objective of this study was to develop a reproducible animal model and thereby to examine the impact of these endocrine-like autoantibodies alone and with thyroid hormone on induction of thyroid-associated atrial tachyarrhythmias. Five New Zealand white rabbits were coimmunized with peptides from the second extracellular loops of the ß1-adrenergic and M2 muscarinic receptors to produce both sympathomimetic and parasympathomimetic antibodies. A catheter-based electrophysiological study was performed on anesthetized rabbits before and after immunization and subsequent treatment with thyroid hormone. Antibody expression facilitated the induction of sustained sinus, junctional and atrial tachycardias, but not AF. Addition of excessive thyroid hormone resulted in induced sustained AF in all animals. AF induction was blocked acutely by the neutralization of these antibodies with immunogenic peptides despite continued hyperthyroidism. The measured atrial effective refractory period as one parameter of AF propensity shortened significantly after immunization and was acutely reversed by peptide neutralization. No further decrease in the effective refractory period was observed after the addition of thyroid hormone, suggesting other cardiac effects of thyroid hormone may contribute to its role in AF induction. This study demonstrates autonomic autoantibodies and thyroid hormone potentiate the vulnerability of the heart to AF, which can be reversed by decoy peptide therapy. These data help fulfill Witebsky's postulates for an increased autoimmune/endocrine basis for Graves' hyperthyroidism and AF.


Subject(s)
Atrial Fibrillation/etiology , Disease Models, Animal , Graves Disease/physiopathology , Receptor, Muscarinic M2/metabolism , Receptors, Adrenergic, beta-1/metabolism , Tachycardia/etiology , Thyroxine/metabolism , Adrenergic beta-1 Receptor Agonists/blood , Adrenergic beta-1 Receptor Agonists/chemistry , Adrenergic beta-1 Receptor Agonists/metabolism , Animals , Antigens/pharmacology , Antigens/therapeutic use , Antigens/toxicity , Atrial Fibrillation/chemically induced , Atrial Fibrillation/immunology , Atrial Fibrillation/prevention & control , Autoantibodies/analysis , Autoantibodies/biosynthesis , Autoantibodies/chemistry , Coronary Sinus/drug effects , Coronary Sinus/immunology , Coronary Sinus/physiopathology , Graves Disease/blood , Graves Disease/immunology , Graves Disease/metabolism , Heart Atria/drug effects , Heart Atria/immunology , Heart Atria/physiopathology , Heart Conduction System/drug effects , Heart Conduction System/immunology , Heart Conduction System/physiopathology , Male , Muscarinic Agonists/blood , Muscarinic Agonists/chemistry , Muscarinic Agonists/metabolism , Peptide Fragments/pharmacology , Peptide Fragments/therapeutic use , Peptide Fragments/toxicity , Rabbits , Receptor, Muscarinic M2/agonists , Receptor, Muscarinic M2/chemistry , Receptors, Adrenergic, beta-1/chemistry , Refractory Period, Electrophysiological/drug effects , Tachycardia/chemically induced , Thyroxine/blood , Thyroxine/pharmacology , Thyroxine/poisoning , Up-Regulation/drug effects
7.
Arthritis Res Ther ; 17: 334, 2015 Nov 21.
Article in English | MEDLINE | ID: mdl-26590032

ABSTRACT

INTRODUCTION: Interleukin-6 (IL-6) is an important mediator of inflammation. In addition to cells involved in inflammation, sensory nociceptive neurons express the IL-6 signal-transducer glycoprotein 130 (gp130). These neurons are not only involved in pain generation but also produce neurogenic inflammation by release of neuropeptides such as calcitonin gene-related peptide (CGRP). Whether IL-6 activation of sensory neurons contributes to the induction of inflammation is unknown. This study explored whether the action of IL-6 on sensory neurons plays a role in the generation of neurogenic inflammation and arthritis induction. METHODS: In SNS-gp130(-/-) mice lacking gp130 selectively in sensory neurons and appropriate control littermates (SNS-gp130(flox/flox)), we induced antigen-induced arthritis (AIA), and assessed swelling, histopathological arthritis scores, pain scores, expression of CGRP in sensory neurons, serum concentrations of CGRP and cytokines, and the cytokine release from single cell suspensions from lymph nodes and spleens. In wild-type mice CGRP release was determined during development of AIA and, in cultured sensory neurons, upon IL-6 stimulation. RESULTS: Compared to SNS-gp130(flox/flox) mice SNS-gp130(-/-) mice showed significantly weaker initial swelling, reduced serum concentrations of CGRP, IL-6, and IL-2, no inflammation-evoked upregulation of CGRP in sensory neurons, but similar histopathological arthritis scores during AIA. During the initial swelling phase of AIA, CGRP was significantly increased in the serum, knee and spleen. In vitro, IL-6 augmented the release of CGRP from cultured sensory neurons. Upon antigen-specific restimulation lymphocytes from SNS-gp130(-/-) mice released more interleukin-17 and interferon-γ than lymphocytes from SNS-gp130(flox/flox) mice. In naive lymphocytes from SNS-gp130(flox/flox) and SNS-gp130(-/-) mice CGRP reduced the release of IL-2 (a cytokine which inhibits the release of interleukin-17 and interferon-γ). CONCLUSIONS: IL-6 signaling in sensory neurons plays a role in the expression of arthritis. Selective deletion of gp130 signaling in sensory neurons reduces the swelling of the joint (most likely by reducing neurogenic inflammation) but increases some proinflammatory systemic cellular responses such as the release of interleukin-17 and interferon-γ from lymphocytes upon antigen-specific restimulation. Thus IL-6 signaling in sensory neurons is not only involved in pain generation but also in the coordination of the inflammatory response.


Subject(s)
Arthritis, Experimental/metabolism , Interleukin-6/pharmacology , Nociceptors/drug effects , Nociceptors/metabolism , Animals , Antigens/toxicity , Arthritis, Experimental/chemically induced , Arthritis, Experimental/pathology , Cytokine Receptor gp130/deficiency , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nociceptors/pathology
8.
Immunology ; 144(1): 107-15, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24995892

ABSTRACT

IgE is known to enhance some antibody responses to specific antigens, but whether this contributes to allergic asthma remains unclear. We have previously found that repeated antigen challenges in mice sensitized with antigen-specific IgE monoclonal antibody (mAb) exacerbated airway inflammation and remodelling accompanied by increased levels of endogenous antigen-specific IgE and IgG1. Here, we investigated whether IgE/antigen-mediated enhancement of endogenous IgE production contributes to the exacerbation of airway inflammation and remodelling. BALB/c mice passively sensitized with ovalbumin (OVA) -specific IgE mAb were challenged with OVA intratracheally seven times; anti-IgE mAb was intraperitoneally administered 1 day before the fourth challenge. Treatment with anti-IgE mAb inhibited the increased level of endogenous OVA-specific IgE in serum, but not OVA-specific IgG1, and a biphasic increase in airway resistance at the fourth challenge. Furthermore, a biphasic increase in airway resistance, airway hyper-responsiveness to methacholine, OVA-specific IgE and IgG1 production, and infiltrations by neutrophils and eosinophils in the lungs at the seventh challenge were suppressed by treatment; airway remodelling, such as goblet cell hyperplasia and sub-epithelial fibrosis, was also reduced. In addition, the production of interleukin-17A, interleukin-33 and CXCL1 in the lungs related to these IgE-mediated responses was decreased by treatment. Collectively, we found that the mechanism leading to the exacerbation of allergic asthma is closely related to IgE/antigen-mediated enhancement of IgE production, suggesting that this may create a vicious circle leading to the chronic status in asthmatic patients having levels of antigen-specific IgE ready to form complexes with antigen.


Subject(s)
Airway Remodeling/immunology , Antigen-Antibody Complex/immunology , Asthma/immunology , Immunoglobulin E/immunology , Lung/immunology , Airway Remodeling/drug effects , Animals , Antibodies, Monoclonal, Murine-Derived/immunology , Antibodies, Monoclonal, Murine-Derived/pharmacology , Antigens/immunology , Antigens/toxicity , Asthma/pathology , Cytokines/immunology , Immunoglobulin G/immunology , Lung/pathology , Male , Mice , Mice, Inbred BALB C , Ovalbumin/immunology , Ovalbumin/toxicity
9.
Proc Natl Acad Sci U S A ; 111(22): 7952-7, 2014 Jun 03.
Article in English | MEDLINE | ID: mdl-24843117

ABSTRACT

Evidence indicates that the densely cultivated region of northeastern China acts as a source for the wind-borne agent of Kawasaki disease (KD). KD is an acute, coronary artery vasculitis of young children, and still a medical mystery after more than 40 y. We used residence times from simulations with the flexible particle dispersion model to pinpoint the source region for KD. Simulations were generated from locations spanning Japan from days with either high or low KD incidence. The postepidemic interval (1987-2010) and the extreme epidemics (1979, 1982, and 1986) pointed to the same source region. Results suggest a very short incubation period (<24 h) from exposure, thus making an infectious agent unlikely. Sampling campaigns over Japan during the KD season detected major differences in the microbiota of the tropospheric aerosols compared with ground aerosols, with the unexpected finding of the Candida species as the dominant fungus from aloft samples (54% of all fungal strains). These results, consistent with the Candida animal model for KD, provide support for the concept and feasibility of a windborne pathogen. A fungal toxin could be pursued as a possible etiologic agent of KD, consistent with an agricultural source, a short incubation time and synchronized outbreaks. Our study suggests that the causative agent of KD is a preformed toxin or environmental agent rather than an organism requiring replication. We propose a new paradigm whereby an idiosyncratic immune response, influenced by host genetics triggered by an environmental exposure carried on winds, results in the clinical syndrome known as acute KD.


Subject(s)
Antigens/toxicity , Edible Grain/toxicity , Environmental Exposure/adverse effects , Mucocutaneous Lymph Node Syndrome/epidemiology , Mucocutaneous Lymph Node Syndrome/etiology , Wind , Agriculture , Antigens/genetics , Antigens, Fungal/genetics , Antigens, Fungal/toxicity , Aspergillus/genetics , Candida/genetics , China/epidemiology , Environmental Exposure/statistics & numerical data , Epidemics/statistics & numerical data , Humans , Incidence , Japan/epidemiology , Models, Statistical , RNA, Ribosomal, 18S/genetics , Vasculitis/epidemiology , Vasculitis/etiology
10.
J Endocrinol ; 219(3): 183-93, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24029729

ABSTRACT

Graves' disease (GD) is a common organ-specific autoimmune disease with the prevalence between 0.5 and 2% in women. Several lines of evidence indicate that the shed A-subunit rather than the full-length thyrotropin receptor (TSHR) is the autoantigen that triggers autoimmunity and leads to hyperthyroidism. We have for the first time induced GD in female rhesus monkeys, which exhibit greater similarity to patients with GD than previous rodent models. After final immunization, the monkeys injected with adenovirus expressing the A-subunit of TSHR (A-sub-Ad) showed some characteristics of GD. When compared with controls, all the test monkeys had significantly higher TSHR antibody levels, half of them had increased total thyroxine (T4) and free T4, and 50% developed goiter. To better understand the underlying mechanisms, quantitative studies on subpopulations of CD4+T helper cells were carried out. The data indicated that this GD model involved a mixed Th1 and Th2 response. Declined Treg proportions and increased Th17:Treg ratio are also observed. Our rhesus monkey model successfully mimicked GD in humans in many aspects. It would be a useful tool for furthering our understanding of the pathogenesis of GD and would potentially shorten the distance toward the prevention and treatment of this disease in human.


Subject(s)
Disease Models, Animal , Graves Disease/physiopathology , Macaca mulatta , Thyroid Gland/physiopathology , Animals , Antigens/genetics , Antigens/toxicity , Autoantibodies/analysis , Biomarkers/blood , Female , Gene Transfer Techniques , Graves Disease/etiology , Graves Disease/immunology , Graves Disease/pathology , Humans , Immunotoxins/genetics , Immunotoxins/toxicity , Organ Size , Protein Subunits/genetics , Protein Subunits/toxicity , Receptors, Thyrotropin/administration & dosage , Receptors, Thyrotropin/genetics , Recombinant Proteins/toxicity , Th1 Cells/immunology , Th1 Cells/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism , Thyroid Gland/immunology , Thyroid Gland/pathology , Thyroxine/blood , Thyroxine/metabolism
11.
J Immunol ; 190(3): 913-21, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23275606

ABSTRACT

We re-examined the observation that γδ T cells, when transferred from mice tolerized to an inhaled conventional Ag, suppress the allergic IgE response to this Ag specifically. Using OVA and hen egg lysozyme in crisscross fashion, we confirmed the Ag-specific IgE-regulatory effect of the γδ T cells. Although only Vγ4(+) γδ T cells are regulators, the Ag specificity does not stem from specificity of their γδ TCRs. Instead, the Vγ4(+) γδ T cells failed to respond to either Ag, but rapidly acquired Ag-specific regulatory function in vivo following i.v. injection of non-T cells derived from the spleen of Ag-tolerized mice. This correlated with their in vivo Ag acquisition from i.v. injected Ag-loaded splenic non-T cells, and in vivo transfer of membrane label provided evidence for direct contact between the injected splenic non-T cells and the Vγ4(+) γδ T cells. Together, our data suggest that Ag itself, when acquired by γδ T cells, directs the specificity of their IgE suppression.


Subject(s)
Antigens/immunology , Asthma/immunology , Immunoglobulin E/immunology , Muramidase/immunology , Ovalbumin/immunology , Receptors, Antigen, T-Cell, gamma-delta/analysis , T-Lymphocyte Subsets/immunology , Administration, Inhalation , Adoptive Transfer , Aerosols , Animals , Antigens/administration & dosage , Antigens/toxicity , Asthma/etiology , Cell Separation , Female , Humans , Immune Tolerance , Immunological Synapses , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Muramidase/administration & dosage , Muramidase/toxicity , Ovalbumin/administration & dosage , Ovalbumin/toxicity , Spleen/immunology , T-Cell Antigen Receptor Specificity , T-Lymphocyte Subsets/transplantation
12.
J Immunol ; 188(7): 3506-12, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22387551

ABSTRACT

Protein arginine methyltransferases (PRMTs), catalyzing methylation of both histones and other cellular proteins, have emerged as key regulators of various cellular processes. This study aimed to identify key PRMTs involved in Ag-induced pulmonary inflammation (AIPI), a rat model for asthma, and to explore the role of PRMT1 in the IL-4-induced eosinophil infiltration process. E3 rats were i.p. sensitized with OVA/alum and intranasally challenged with OVA to induce AIPI. The expressions of PRMT1-6, eotaxin-1, and CCR3 in lungs were screened by real-time quantitative PCR. Arginine methyltransferase inhibitor 1 (AMI-1, a pan-PRMT inhibitor) and small interfering RNA-PRMT1 were used to interrupt the function of PRMT1 in A549 cells. In addition, AMI-1 was administrated intranasally to AIPI rats to observe the effects on inflammatory parameters. The results showed that PRMT1 expression was mainly expressed in bronchus and alveolus epithelium and significantly upregulated in lungs from AIPI rats. The inhibition of PRMTs by AMI-1 and the knockdown of PRMT1 expression were able to downregulate the expressions of eotaxin-1 and CCR3 with the IL-4 stimulation in the epithelial cells. Furthermore, AMI-1 administration to AIPI rats can also ameliorate pulmonary inflammation, reduce IL-4 production and humoral immune response, and abrogate eosinophil infiltration into the lungs. In summary, PRMT1 expression is upregulated in AIPI rat lungs and can be stimulated by IL-4. Intervention of PRMT1 activity can abrogate IL-4-dependent eotaxin-1 production to influence the pulmonary inflammation with eosinophil infiltration. The findings may provide experimental evidence that PRMT1 plays an important role in asthma pathogenesis.


Subject(s)
Antigens/toxicity , Chemokine CCL11/biosynthesis , Epithelial Cells/metabolism , Interleukin-4/pharmacology , Protein-Arginine N-Methyltransferases/physiology , Pulmonary Eosinophilia/immunology , Animals , Asthma/metabolism , Chemokine CCL11/genetics , Disease Models, Animal , Drug Evaluation, Preclinical , Epithelial Cells/drug effects , Female , Gene Expression Regulation/drug effects , Male , Naphthalenesulfonates/pharmacology , Naphthalenesulfonates/therapeutic use , Ovalbumin/toxicity , Protein-Arginine N-Methyltransferases/antagonists & inhibitors , Protein-Arginine N-Methyltransferases/biosynthesis , Protein-Arginine N-Methyltransferases/genetics , Pulmonary Eosinophilia/chemically induced , Pulmonary Eosinophilia/drug therapy , Pulmonary Eosinophilia/enzymology , RNA Interference , RNA, Small Interfering/pharmacology , Rats , Recombinant Proteins/pharmacology , Respiratory System/cytology , Specific Pathogen-Free Organisms , Urea/analogs & derivatives , Urea/pharmacology , Urea/therapeutic use
13.
Biol Pharm Bull ; 34(12): 1835-40, 2011.
Article in English | MEDLINE | ID: mdl-22130239

ABSTRACT

The development of a simple, easy-to-use, and non-invasive vaccination system is in high demand. For transcutaneous immunization (TCI), we previously developed a hydrogel patch formulation that accelerates the penetration of an antigen (Ag) through the stratum corneum (SC) and effectively elicits Ag-specific immune responses. The present studies were performed to optimize the composition and assess the safety of the patch formulation. A hydrogel patch with a water content ratio of 5% more effectively induced an immune response compared to patches with a different composition, suggesting that the moisture content of the hydrogel patch formulation has optimal ratio for SC hydration to promote Ag penetration through the SC. TCI using a hydrogel patch induced few local and systemic adverse reactions. Based on these results, we are now advancing translational research to evaluate the safety and efficacy of our novel TCI system in humans.


Subject(s)
Antigens/administration & dosage , Hydrogel, Polyethylene Glycol Dimethacrylate/administration & dosage , Transdermal Patch , Vaccination/methods , Administration, Cutaneous , Animals , Antigens/toxicity , Diphtheria Toxin/administration & dosage , Diphtheria Toxin/toxicity , Female , Humans , Hydrogel, Polyethylene Glycol Dimethacrylate/toxicity , In Vitro Techniques , Male , Ovalbumin/administration & dosage , Ovalbumin/toxicity , Rats , Rats, Hairless , Rats, Wistar , Skin/drug effects , Skin Tests , Tetanus Toxin/administration & dosage , Tetanus Toxin/toxicity , Transdermal Patch/adverse effects
14.
Toxins (Basel) ; 3(9): 1146-62, 2011 09.
Article in English | MEDLINE | ID: mdl-22069760

ABSTRACT

Enterotoxigenic Escherichia coli (ETEC) strains are a major cause of diarrhea in humans and animals. Heat-stable (STa) and heat-labile (LT) enterotoxins produced by ETEC disrupt fluid homeostasis in host small intestinal epithelial cells and cause fluid and electrolyte hyper-secretion that leads to diarrhea. ETEC strains producing STa or LT are sufficiently virulent to cause diarrhea, therefore STa and LT antigens must be included in ETEC vaccines. However, potent toxicity and poor immunogenicity (of STa) prevent them from being directly applied as vaccine components. While LT toxoids, especially LT(R192G), being used in vaccine development, STa toxoids have not been included. A recent study (IAI, 78:316-325) demonstrated porcine-type STa toxoids [pSTa(P12F) and pSTa(A13Q)] elicited protective anti-STa antibodies after being fused to a porcine-type LT toxoid [pLT(R192G)]. In this study, we substituted the 8th, 9th, 16th, or the 17th amino acid of a human-type STa (hSTa) and generated 28 modified STa peptides. We tested each STa peptide for toxicity and structure integrity, and found nearly all modified STa proteins showed structure alteration and toxicity reduction. Based on structure similarity and toxic activity, three modified STa peptides: STa(E8A), STa(T16Q) and STa(G17S), were selected to construct LT(192)-STa(-toxoid) fusions. Constructed fusions were used to immunize mice, and immunized mice developed anti-STa antibodies. Results from this study provide useful information in developing toxoid vaccines against ETEC diarrhea.


Subject(s)
Antigens/immunology , Bacterial Toxins/immunology , Enterotoxins/immunology , Escherichia coli Proteins/immunology , Recombinant Fusion Proteins/immunology , Animals , Antibodies/immunology , Antigens/genetics , Antigens/toxicity , Bacterial Toxins/genetics , Bacterial Toxins/toxicity , Enterotoxigenic Escherichia coli , Enterotoxins/genetics , Enterotoxins/toxicity , Escherichia coli Infections/prevention & control , Escherichia coli Proteins/genetics , Escherichia coli Proteins/toxicity , Escherichia coli Vaccines/administration & dosage , Female , Intestinal Mucosa/metabolism , Intestines/drug effects , Mice , Mice, Inbred BALB C , Peptides/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/toxicity , Swine
15.
Int J Pharm ; 416(2): 486-92, 2011 Sep 20.
Article in English | MEDLINE | ID: mdl-21382458

ABSTRACT

UNLABELLED: Encapsulation of glucocorticoids into long-circulating liposomes provides targeting of these drugs to the inflamed synovium in experimental arthritis and thereby strongly improves their therapeutic index. The goal of this study was to evaluate the effect and mechanisms of intravenous liposomal delivery of prednisolone phosphate (Lip-PLP) on protease mediated cartilage destruction during murine antigen-induced arthritis (AIA). Mice treated with a single injection of Lip-PLP showed a pronounced suppression of synovial immune cell infiltration compared to control, PBS-treated mice. Liposomal PLP also significantly suppressed interleukin 1ß (3.6 fold) in the synovium, but not in the blood serum. Furthermore, expression of the proteases MMP-3, -9, -13 and -14 and ADAMTS-4 and -5 was suppressed by Lip-PLP in the synovium, but not within the articular cartilage of the femur and tibia, demonstrating the specific action of Lip-PLP on the synovium. Lip-PLP is phagocytosed by macrophages in vitro and suppresses their expression of IL-1ß and MMPs after LPS activation. Moreover, Lip-PLP suppresses destruction of the cartilage matrix during AIA mediated by active MMPs and ADAMTS, as assessed by immunostaining of their respective neoepitopes VDIPEN and NITEGE in various cartilage layers of the knee joint. CONCLUSION: liposomal delivery of glucocorticoids protects against cartilage matrix destruction during experimental arthritis by inhibiting protease expression and activity in the inflamed synovium.


Subject(s)
Arthritis, Experimental/drug therapy , Drug Delivery Systems , Glucocorticoids/pharmacology , Prednisolone/analogs & derivatives , ADAM Proteins/drug effects , ADAM Proteins/metabolism , Animals , Antigens/toxicity , Arthritis, Experimental/pathology , Cartilage, Articular/drug effects , Cartilage, Articular/pathology , Delayed-Action Preparations , Glucocorticoids/administration & dosage , Interleukin-1beta/metabolism , Liposomes , Male , Matrix Metalloproteinases/metabolism , Mice , Mice, Inbred C57BL , Prednisolone/administration & dosage , Prednisolone/pharmacology , Synovial Membrane/drug effects , Synovial Membrane/pathology
16.
Int J Pharm ; 416(2): 493-8, 2011 Sep 20.
Article in English | MEDLINE | ID: mdl-21382459

ABSTRACT

UNLABELLED: The goal of this study was to compare the effects of liposomal and free glucocorticoid formulations on joint inflammation and activity of the hypothalamic-pituitary-adrenal (HPA) axis during experimental antigen-induced arthritis (AIA). A dose of 10mg/kg liposomal prednisolone phosphate (PLP) gave a suppression of the HPA-axis, as measured by plasma corticosterone levels in mice with AIA and in naïve mice. In a subsequent dose-response study, we found that a single dose of 1mg/kg liposomal prednisolone phosphate (PLP) was still equally effective in suppressing joint inflammation as 4 repeated once-daily injections of 10mg/kg free PLP. Moreover, the 1mg/kg liposomal PLP dose gave 22% less suppression of corticosterone levels than 10mg/kg of liposomal PLP at day 14 of the AIA. In order to further optimize liposomal glucocorticoids, we compared liposomal PLP with liposomal budesonide phosphate (BUP) (1mg/kg). At 1 day after treatment, liposomal BUP gave a significantly stronger suppression of joint swelling than liposomal PLP (lip. BUP 98% vs. lip. PLP 79%). Both formulations also gave a strong and lasting suppression of synovial infiltration in equal amounts. However, at day 21 after AIA, liposomal PLP still significantly suppressed corticosterone levels, whereas this suppression was not longer statistically significant for liposomal BUP. CONCLUSION: Liposomal delivery improves the safety of glucocorticoids by allowing for lower effective dosing. The safety of liposomal glucocorticoid may be further improved by encapsulating BUP rather than PLP.


Subject(s)
Arthritis, Experimental/drug therapy , Budesonide/administration & dosage , Glucocorticoids/administration & dosage , Prednisolone/analogs & derivatives , Animals , Antigens/toxicity , Arthritis, Experimental/pathology , Budesonide/pharmacology , Budesonide/toxicity , Corticosterone/blood , Dose-Response Relationship, Drug , Glucocorticoids/pharmacology , Glucocorticoids/toxicity , Liposomes , Male , Mice , Mice, Inbred C57BL , Prednisolone/administration & dosage , Prednisolone/pharmacology , Prednisolone/toxicity , Synovial Membrane/drug effects , Synovial Membrane/pathology
17.
Allergy ; 66(4): 523-31, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21091718

ABSTRACT

BACKGROUND: Adult bone marrow-derived mesenchymal stem cells (MSC) possess potent immune modulatory effects which support their possible use as a therapy for immune-mediated disease. MSC induce regulatory T cells (T(reg)) in vitro although the in vivo relevance of this is not clear. OBJECTIVE: This study addressed the hypothesis that adult bone marrow derived-MSC would prevent the pathology associated with allergen-driven airway inflammation, and sought to define the effector mechanism. METHODS: The influence of allogeneic MSC was examined in a model system where T(reg) induction is essential to prevent pathology. This was tested using a combination of a model of ovalbumin-driven inflammation with allogeneic MSC cell therapy. RESULTS: Systemic administration of allogeneic MSC protected the airways from allergen-induced pathology, reducing airway inflammation and allergen-specific IgE. MSC were not globally suppressive but induced CD4(+) FoxP3(+) T cells and modulated cell-mediated responses at a local and systemic level, decreasing IL-4 but increasing IL-10 in bronchial fluid and from allergen re-stimulated splenocytes. Moderate dose cyclophosphamide protocols were used to differentially ablate T(reg) responses; under these conditions the major beneficial effect of MSC therapy was lost, suggesting induction of T(reg) as the key mechanism of action by MSC in this model. In spite of the elimination of T(reg) , a significant reduction in airway eosinophilia persisted in those treated with MSC. CONCLUSION: These data demonstrate that MSC induce T(reg) in vivo and reduce allergen-driven pathology. Multiple T(reg) dependent and independent mechanisms of therapeutic action are employed by MSC.


Subject(s)
Bronchial Hyperreactivity/prevention & control , Inflammation/prevention & control , Mesenchymal Stem Cell Transplantation , T-Lymphocytes, Regulatory/immunology , Animals , Antigens/immunology , Antigens/toxicity , Bronchial Hyperreactivity/immunology , Female , Inflammation/immunology , Mesenchymal Stem Cells/immunology , Mice , Mice, Inbred BALB C , Ovalbumin/immunology , Ovalbumin/toxicity
18.
J Immunol ; 183(6): 4003-12, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19717512

ABSTRACT

Because IL-1beta plays an important role in inflammation in human and murine arthritis, we investigated the contribution of the inflammasome components ASC, NALP-3, IPAF, and caspase-1 to inflammatory arthritis. We first studied the phenotype of ASC-deficient and wild-type mice during Ag-induced arthritis (AIA). ASC(-/-) mice showed reduced severity of AIA, decreased levels of synovial IL-1beta, and diminished serum amyloid A levels. In contrast, mice deficient in NALP-3, IPAF, or caspase-1 did not show any alteration of joint inflammation, thus indicating that ASC associated effects on AIA are independent of the classical NALP-3 or IPAF inflammasomes. Because ASC is a ubiquitous cytoplasmic protein that has been implicated in multiple cellular processes, we explored other pathways through which ASC may modulate inflammation. Ag-specific proliferation of lymph node and spleen cells from ASC-deficient mice was significantly decreased in vitro, as was the production of IFN-gamma, whereas IL-10 production was enhanced. TCR ligation by anti-CD3 Abs in the presence or absence of anti-CD28 Abs induced a reduction in T cell proliferation in ASC(-/-) T cells compared with wild-type ones. In vivo lymph node cell proliferation was also significantly decreased in ASC(-/-) mice, but no effects on apoptosis were observed either in vitro or in vivo in these mice. In conclusion, these results strongly suggest that ASC modulates joint inflammation in AIA through its effects on cell-mediated immune responses but not via its implication in inflammasome formation.


Subject(s)
Apoptosis Regulatory Proteins/deficiency , Arthritis, Experimental/etiology , Calcium-Binding Proteins/deficiency , Carrier Proteins/genetics , Caspase 1/deficiency , Cytoskeletal Proteins/physiology , Inflammation/etiology , Animals , Antigens/toxicity , Arthritis, Experimental/pathology , CARD Signaling Adaptor Proteins , Cell Proliferation , Joint Diseases/pathology , Lymph Nodes/pathology , Mice , Mice, Knockout , Multiprotein Complexes/immunology , NLR Family, Pyrin Domain-Containing 3 Protein , Spleen/pathology
19.
Mucosal Immunol ; 2(5): 427-38, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19571798

ABSTRACT

The role of CD8(+) T cells in oral tolerance remains unclear. To address this, we developed a model to induce CD8(+) Tregs by feeding the major histocompatibility complex class I immunodominant epitope of OVA, OVA((257-264)). OVA((257-264)) feeding induced tolerance similar to that observed in OVA protein-fed mice, capable of suppressing the production of Th1 and Th17 cytokines and inhibiting a Th1-driven delayed-type hypersensitivity response following immunization with whole OVA (wOVA) protein. OVA((257-264)) peptide-induced suppression could be transferred to naive mice with CD8(+) cells, but not CD8-depleted cells, isolated from mesenteric lymph nodes of peptide-fed mice. Interestingly, while capable of inhibiting Th1 and Th17 responses, OVA((257-264)) feeding could not suppress any feature of a Th2 inflammatory response, though OVA protein feeding could, suggesting that these cells function through a different mechanism than their CD4(+) counterparts generated in response to feeding with wOVA. Thus, CD8(+) T cells are functionally capable of mediating tolerance to Th1 and Th17 responses.


Subject(s)
Alveolitis, Extrinsic Allergic/prevention & control , Antigens/administration & dosage , CD8-Positive T-Lymphocytes/immunology , Desensitization, Immunologic/methods , Immune Tolerance/immunology , Immunodominant Epitopes/administration & dosage , Ovalbumin/administration & dosage , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Regulatory/immunology , Administration, Intranasal , Administration, Oral , Adoptive Transfer , Alveolitis, Extrinsic Allergic/immunology , Alveolitis, Extrinsic Allergic/pathology , Animals , Antigens/immunology , Antigens/toxicity , CD8-Positive T-Lymphocytes/transplantation , Ear, External/immunology , Ear, External/pathology , Edema/etiology , Edema/immunology , Edema/pathology , Immunization , Immunodominant Epitopes/immunology , Immunodominant Epitopes/toxicity , Injections, Intradermal , Lymphocyte Cooperation , Mice , Mice, Inbred C57BL , Models, Immunological , Ovalbumin/immunology , Ovalbumin/toxicity , Peptide Fragments/administration & dosage , Peptide Fragments/immunology , Peptide Fragments/toxicity , T-Lymphocyte Subsets/immunology
20.
Arthritis Res Ther ; 10(6): R133, 2008.
Article in English | MEDLINE | ID: mdl-19014513

ABSTRACT

INTRODUCTION: Adrenomedullin is a potent vasodilatory and hypotensive peptide as well as an endogenous immunomodulatory factor with predominantly anti-inflammatory effects. The purpose of the present study was to evaluate the therapeutic effects of adrenomedullin in rabbits with antigen-induced arthritis, an experimental model of rheumatoid arthritis. METHODS: Following the induction of arthritis in both knee joints by ovalbumin injection into the joint spaces of pre-immunized rabbits, increasing daily doses of adrenomedullin were injected into the knee joint spaces or saline was injected into the contralateral knee joint spaces as the control. For time-course experiments, adrenomedullin and saline were injected into the knee joint spaces daily for 7 days and 20 days. The degree of joint swelling and the histological change in the knee joints injected with adrenomedullin were compared with the control knee joints. Histological evaluation of the infrapatellar fat pads and synovial tissue was performed. TNFalpha, IL-6, vascular endothelial growth factor and transforming growth factor-beta mRNA levels in the synovial tissue were measured using real-time quantitative PCR. RESULTS: Daily injections of adrenomedullin into the knee joint spaces of rabbits with antigen-induced arthritis decreased joint swelling. Histological examination revealed that adrenomedullin reduced edematous changes and the infiltration of inflammatory cells in the synovial tissues. Analysis of mRNA levels showed that adrenomedullin significantly reduced TNFalpha mRNA expression by 21% to 49% in a dose-dependent manner, and dose-dependently increased IL-6 mRNA expression by 45% to 121%. CONCLUSIONS: These results suggest that daily injections of adrenomedullin into the knee joint spaces of rabbits with antigen-induced arthritis ameliorated the inflammatory response in arthritic joints. Adrenomedullin may thus be useful as a treatment for rheumatoid arthritis; however, the effect of adrenomedullin on IL-6 production in the synovial tissue may be an undesirable adverse effect in rheumatoid arthritis therapy.


Subject(s)
Adrenomedullin/administration & dosage , Arthritis, Experimental/drug therapy , Ovalbumin/toxicity , Animals , Antigens/toxicity , Arthritis, Experimental/chemically induced , Arthritis, Experimental/pathology , Female , Injections, Intra-Articular , Rabbits
SELECTION OF CITATIONS
SEARCH DETAIL
...