Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Immunity ; 29(1): 79-89, 2008 Jul 18.
Article in English | MEDLINE | ID: mdl-18571443

ABSTRACT

DR3 (TRAMP, LARD, WSL-1, TNFRSF25) is a death-domain-containing tumor necrosis factor (TNF)-family receptor primarily expressed on T cells. TL1A, the TNF-family ligand for DR3, can costimulate T cells, but the physiological function of TL1A-DR3 interactions in immune responses is not known. Using DR3-deficient mice, we identified DR3 as the receptor responsible for TL1A-induced T cell costimulation and dendritic cells as the likely source for TL1A during T cell activation. Despite its role in costimulation, DR3 was not required for in vivo T cell priming, for polarization into T helper 1 (Th1), Th2, or Th17 effector cell subtypes, or for effective control of infection with Toxoplasma gondii. Instead, DR3 expression was required on T cells for immunopathology, local T cell accumulation, and cytokine production in Experimental Autoimmune Encephalomyelitis (EAE) and allergic lung inflammation, disease models that depend on distinct effector T cell subsets. DR3 could be an attractive therapeutic target for T cell-mediated autoimmune and allergic diseases.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Inflammation/immunology , Lymphocyte Activation/immunology , Receptors, Tumor Necrosis Factor, Member 25/immunology , T-Lymphocyte Subsets/immunology , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , Cell Proliferation , Encephalomyelitis, Autoimmune, Experimental/immunology , Inflammation/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Tumor Necrosis Factor, Member 25/metabolism , Respiratory Hypersensitivity/immunology , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/metabolism , Toxoplasmosis/immunology , Tumor Necrosis Factor Ligand Superfamily Member 15/immunology , Tumor Necrosis Factor Ligand Superfamily Member 15/metabolism
2.
J Immunol ; 194(8): 3567-82, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25786692

ABSTRACT

The TNF family cytokine TL1A (Tnfsf15) costimulates T cells and type 2 innate lymphocytes (ILC2) through its receptor DR3 (Tnfrsf25). DR3-deficient mice have reduced T cell accumulation at the site of inflammation and reduced ILC2-dependent immune responses in a number of models of autoimmune and allergic diseases. In allergic lung disease models, immunopathology and local Th2 and ILC2 accumulation is reduced in DR3-deficient mice despite normal systemic priming of Th2 responses and generation of T cells secreting IL-13 and IL-4, prompting the question of whether TL1A promotes the development of other T cell subsets that secrete cytokines to drive allergic disease. In this study, we find that TL1A potently promotes generation of murine T cells producing IL-9 (Th9) by signaling through DR3 in a cell-intrinsic manner. TL1A enhances Th9 differentiation through an IL-2 and STAT5-dependent mechanism, unlike the TNF-family member OX40, which promotes Th9 through IL-4 and STAT6. Th9 differentiated in the presence of TL1A are more pathogenic, and endogenous TL1A signaling through DR3 on T cells is required for maximal pathology and IL-9 production in allergic lung inflammation. Taken together, these data identify TL1A-DR3 interactions as a novel pathway that promotes Th9 differentiation and pathogenicity. TL1A may be a potential therapeutic target in diseases dependent on IL-9.


Subject(s)
Asthma/immunology , Cell Differentiation/immunology , Interleukin-9/immunology , Receptors, Tumor Necrosis Factor, Member 25/immunology , T-Lymphocytes, Helper-Inducer/immunology , Tumor Necrosis Factor Ligand Superfamily Member 15/immunology , Animals , Asthma/genetics , Asthma/pathology , Cell Differentiation/genetics , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Interleukin-13/genetics , Interleukin-13/immunology , Interleukin-4/genetics , Interleukin-4/immunology , Interleukin-9/genetics , Mice , Mice, Knockout , Receptors, Tumor Necrosis Factor, Member 25/genetics , Signal Transduction/genetics , Signal Transduction/immunology , T-Lymphocytes, Helper-Inducer/pathology , Tumor Necrosis Factor Ligand Superfamily Member 15/genetics
3.
Mol Pharm ; 10(8): 2849-57, 2013 Aug 05.
Article in English | MEDLINE | ID: mdl-23768126

ABSTRACT

To enhance the immune activity of vaccine adjuvants polyinosinic:polycytidylic acid (poly I:C) and CpG acetalated dextran (Ac-DEX) microparticles can be used. Ac-DEX is a biodegradable and water-insoluble polymer that degrades significantly faster at pH 5.0 (phagosomal pH) than at pH 7.4 and has tunable degradation rates that can range from hours to months. This is an ideal characteristic for delivery of an antigen and adjuvant within the lysosomal compartment of a phagocytic cell. We evaluated poly I:C and CpG encapsulated in Ac-DEX microparticles using RAW macrophages as a model antigen-presenting cell. These cells were cultured with poly I:C or CpG in their free form, encapsulated in a fast degrading Ac-DEX, in slow degrading Ac-DEX, or in the Food and Drug Administration-approved polymer poly(lactic-co-glycolic acid) (PLGA). Ac-DEX had higher encapsulation efficiencies for both poly I:C and CpG than PLGA. Furthermore, poly I:C or CpG encapsulated in Ac-DEX also showed, in general, a significantly stronger immunostimulatory response than PLGA and unencapsulated CpG or poly I:C, which was indicated by a higher rate of nitric oxide release and increased levels of cytokines such as TNF-α, IL-6, IL-10, and IFN-γ. Overall, we have illustrated a method for enhancing the delivery of these vaccine adjuvants to further enhance the development of Ac-DEX vaccine formulations.


Subject(s)
Dinucleoside Phosphates/metabolism , Poly I-C/metabolism , Toll-Like Receptors/agonists , Animals , Cell Line , Dextrans/chemistry , Macrophages/drug effects , Macrophages/metabolism , Mice , Microscopy, Atomic Force
4.
Pharm Res ; 30(5): 1349-61, 2013 May.
Article in English | MEDLINE | ID: mdl-23354770

ABSTRACT

PURPOSE: A rapid immune response is required to prevent death from Anthrax, caused by Bacillus anthracis. METHOD: We formulated a vaccine carrier comprised of acetalated dextran microparticles encapsulating recombinant protective antigen (rPA) and resiquimod (a toll-like receptor 7/8 agonist). RESULTS: We were able to protect against triplicate lethal challenge by vaccinating twice (Days 0, 7) and then aggressively challenging on Days 14, 21, 28. A significantly higher level of antibodies was generated by day 14 with the encapsulated group compared to the conventional rPA and alum group. Antibodies produced by the co-encapsulated group were only weakly-neutralizing in toxin neutralization; however, survival was not dependent on toxin neutralization, as all vaccine formulations survived all challenges except control groups. Post-mortem culture swabs taken from the hearts of vaccinated groups that did not produce significant neutralizing titers failed to grow B. anthracis. CONCLUSIONS: Results indicate that protective antibodies are not required for rapid protection; indeed, cytokine results indicate that T cell protection may play a role in protection from anthrax. We report the first instance of use of a particulate carrier to generate a rapid protective immunity against anthrax.


Subject(s)
Anthrax Vaccines/therapeutic use , Anthrax/prevention & control , Bacillus anthracis/immunology , Dextrans/chemistry , Drug Carriers/chemistry , Acetylation , Animals , Anthrax/immunology , Anthrax/microbiology , Anthrax Vaccines/administration & dosage , Anthrax Vaccines/immunology , Antibody Formation , Antigens, Bacterial/administration & dosage , Antigens, Bacterial/immunology , Antigens, Bacterial/therapeutic use , Bacterial Toxins/administration & dosage , Bacterial Toxins/immunology , Bacterial Toxins/therapeutic use , Imidazoles/administration & dosage , Imidazoles/therapeutic use , Mice , Toll-Like Receptors/agonists , Vaccination , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology , Vaccines, Subunit/therapeutic use
5.
Proc Natl Acad Sci U S A ; 107(12): 5652-7, 2010 Mar 23.
Article in English | MEDLINE | ID: mdl-20231466

ABSTRACT

Bone marrow stromal cells [BMSCs; also known as mesenchymal stem cells (MSCs)] effectively suppress inflammatory responses in acute graft-versus-host disease in humans and in a number of disease models in mice. Many of the studies concluded that BMSC-driven immunomodulation is mediated by the suppression of proinflammatory Th1 responses while rebalancing the Th1/Th2 ratio toward Th2. In this study, using a ragweed induced mouse asthma model, we studied if BMSCs could be beneficial in an allergic, Th2-dominant environment. When BMSCs were injected i.v. at the time of the antigen challenge, they protected the animals from the majority of asthma-specific pathological changes, including inhibition of eosinophil infiltration and excess mucus production in the lung, decreased levels of Th2 cytokines (IL-4, IL-5, and IL-13) in bronchial lavage, and lowered serum levels of Th2 immunoglobulins (IgG1 and IgE). To explore the mechanism of the effect we used BMSCs isolated from a variety of knockout mice, performed in vivo blocking of cytokines and studied the effect of asthmatic serum and bronchoalveolar lavage from ragweed challenged animals on the BMSCs in vitro. Our results suggest that IL-4 and/or IL-13 activate the STAT6 pathway in the BMSCs resulting in an increase of their TGF-beta production, which seems to mediate the beneficial effect, either alone, or together with regulatory T cells, some of which might be recruited by the BMSCs. These data suggest that, in addition to focusing on graft-versus-host disease and autoimmune diseases, allergic conditions--specifically therapy resistant asthma--might also be a likely target of the recently discovered cellular therapy approach using BMSCs.


Subject(s)
Asthma/immunology , Mesenchymal Stem Cells/immunology , Transforming Growth Factor beta/immunology , Ambrosia/adverse effects , Ambrosia/immunology , Animals , Asthma/etiology , Asthma/pathology , Asthma/therapy , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Cytokines/deficiency , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Humans , Immunosuppression Therapy , In Vitro Techniques , Lung/immunology , Lung/pathology , Mesenchymal Stem Cell Transplantation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Th2 Cells/immunology , Transplantation, Homologous , Transplantation, Isogeneic
6.
Anal Biochem ; 425(1): 54-61, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22406431

ABSTRACT

Rapid presymptomatic diagnosis of Bacillus anthracis at early stages of infection plays a crucial role in prompt medical intervention to prevent rapid disease progression and accumulation of lethal levels of toxin. To detect low levels of the anthrax protective antigen (PA) exotoxin in biological fluids, we have developed a metal-enhanced fluorescence (MEF)-PA assay using a combination of the MEF effect and microwave-accelerated PA protein surface absorption. The assay is based on a modified version of our "rapid catch and signal" (RCS) technology previously designed for the ultra-fast and sensitive analysis of genomic DNA sequences. Technologically, the proposed MEF-PA assay uses standard 96-well plastic plates modified with silver island films (SiFs) grown within the wells. It is shown that the fluorescent probe, covalently attached to the secondary antibody, plays a crucial role of indicating complex formation (i.e., shows a strong MEF response to the recognition event). Microwave irradiation rapidly accelerates PA deposition onto the surface ("rapid catch"), significantly speeding up the MEF-PA assay and resulting in a total assay run time of less than 40 min with an analytical sensitivity of less than 1 pg/ml PA.


Subject(s)
Antigens, Bacterial/analysis , Bacterial Toxins/analysis , Fluorescence , Microwaves , Silver/chemistry , Antigens, Bacterial/chemistry , Bacillus anthracis/immunology , Bacillus anthracis/isolation & purification , Bacterial Toxins/chemistry , Metals/chemistry
8.
Infect Immun ; 79(8): 3012-9, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21576337

ABSTRACT

Bacillus cereus G9241 was isolated from a welder with a pulmonary anthrax-like illness. The organism contains two megaplasmids, pBCXO1 and pBC218. These plasmids are analogous to the Bacillus anthracis Ames plasmids pXO1 and pXO2 that encode anthrax toxins and capsule, respectively. Here we evaluated the virulence of B. cereus G9241 as well as the contributions of pBCXO1 and pBC218 to virulence. B. cereus G9241 was avirulent in New Zealand rabbits after subcutaneous inoculation and attenuated 100-fold compared to the published 50% lethal dose (LD(50)) values for B. anthracis Ames after aerosol inoculation. A/J and C57BL/6J mice were comparably susceptible to B. cereus G9241 by both subcutaneous and intranasal routes of infection. However, the LD(50)s for B. cereus G9241 in both mouse strains were markedly higher than those reported for B. anthracis Ames and more like those of the toxigenic but nonencapsulated B. anthracis Sterne. Furthermore, B. cereus G9241 spores could germinate and disseminate after intranasal inoculation into A/J mice, as indicated by the presence of vegetative cells in the spleen and blood of animals 48 h after infection. Lastly, B. cereus G9241 derivatives cured of one or both megaplasmids were highly attenuated in A/J mice. We conclude that the presence of the toxin- and capsule-encoding plasmids pBCXO1 and pBC218 in B. cereus G9241 alone is insufficient to render the strain as virulent as B. anthracis Ames. However, like B. anthracis, full virulence of B. cereus G9241 for mice requires the presence of both plasmids.


Subject(s)
Anthrax/pathology , Antigens, Bacterial/biosynthesis , Bacillus anthracis/metabolism , Bacillus anthracis/pathogenicity , Bacillus cereus/metabolism , Bacillus cereus/pathogenicity , Bacterial Capsules/biosynthesis , Bacterial Toxins/biosynthesis , Administration, Inhalation , Aerosols/administration & dosage , Animals , Anthrax/microbiology , Antigens, Bacterial/genetics , Bacillus anthracis/genetics , Bacillus cereus/genetics , Bacterial Capsules/genetics , Bacterial Toxins/genetics , Disease Models, Animal , Female , Lethal Dose 50 , Mice , Mice, Inbred A , Mice, Inbred C57BL , Plasmids/analysis , Rabbits , Rodent Diseases/microbiology , Rodent Diseases/pathology , Virulence , Virulence Factors/biosynthesis , Virulence Factors/genetics
9.
J Exp Med ; 202(6): 829-39, 2005 Sep 19.
Article in English | MEDLINE | ID: mdl-16172260

ABSTRACT

Thymic stromal lymphopoietin (TSLP) is a cytokine that promotes CD4+ T cell homeostasis. We now demonstrate that TSLP is required to mount a normal CD4+ T cell-mediated inflammatory response. TSLP acts directly on naive, but not, memory CD4+ T cells, and promotes their proliferation in response to antigen. In addition, TSLP exerts an effect indirectly through DCs to promote Th2 differentiation of CD4+ T cells. Correspondingly, TSLP receptor (TSLPR) knockout (KO) mice exhibit strong Th1 responses, with high levels of interleukin (IL)-12, interferon-gamma, and immunoglobulin (Ig) G2a, but low production of IL-4, -5, -10, -13, and IgE; moreover, CD4+ T cells from these animals proliferate less well in response to antigen. Furthermore, TSLPR KO mice fail to develop an inflammatory lung response to inhaled antigen unless supplemented with wild-type CD4+ T cells. This underscores an important role for this cytokine in the development of inflammatory and/or allergic responses in vivo.


Subject(s)
Asthma/metabolism , Cytokines/physiology , Lung/pathology , Animals , Asthma/pathology , CD4-Positive T-Lymphocytes/metabolism , Cell Proliferation , Cells, Cultured , Dendritic Cells/metabolism , Disease Models, Animal , Immunoglobulins , Inflammation/immunology , Inflammation/metabolism , Interferon-gamma/biosynthesis , Lung/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Receptors, Cytokine/deficiency , Receptors, Cytokine/genetics , Thymic Stromal Lymphopoietin
10.
J Clin Invest ; 117(10): 2941-51, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17885690

ABSTRACT

Development of persistent Th2 responses in asthma and chronic helminth infections are a major health concern. IL-10 has been identified as a critical regulator of Th2 immunity, but mechanisms for controlling Th2 effector function remain unclear. IL-10 also has paradoxical effects on Th2-associated pathology, with IL-10 deficiency resulting in increased Th2-driven inflammation but also reduced airway hyperreactivity (AHR), mucus hypersecretion, and fibrosis. We demonstrate that increased IL-13 receptor alpha 2 (IL-13Ralpha2) expression is responsible for the reduced AHR, mucus production, and fibrosis in BALB/c IL-10(-/-) mice. Using models of allergic asthma and chronic helminth infection, we demonstrate that IL-10 and IL-13Ralpha2 coordinately suppress Th2-mediated inflammation and pathology, respectively. Although IL-10 was identified as the dominant antiinflammatory mediator, studies with double IL-10/IL-13Ralpha2-deficient mice illustrate an indispensable role for IL-13Ralpha2 in the suppression of AHR, mucus production, and fibrosis. Thus, IL-10 and IL-13Ralpha2 are both required to control chronic Th2-driven pathological responses.


Subject(s)
Asthma/genetics , Bronchial Hyperreactivity/genetics , Bronchitis/genetics , Interleukin-10/physiology , Interleukin-13 Receptor alpha2 Subunit/physiology , Th2 Cells/immunology , Animals , Asthma/immunology , Asthma/pathology , Bronchial Hyperreactivity/immunology , Bronchial Hyperreactivity/pathology , Bronchitis/immunology , Bronchitis/pathology , Fibrosis , Granuloma/genetics , Granuloma/immunology , Granuloma/pathology , Interleukin-10/genetics , Interleukin-13 Receptor alpha2 Subunit/genetics , Mice , Mice, Mutant Strains , Mucus/metabolism , Th1 Cells/immunology
11.
Microbiology (Reading) ; 156(Pt 10): 2982-2993, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20688829

ABSTRACT

Many genes in Bacillus cereus and Bacillus thuringiensis are under the control of the transcriptional regulator PlcR and its regulatory peptide, PapR. In Bacillus anthracis, the causative agent of anthrax, PlcR is inactivated by truncation, and consequently genes having PlcR binding sites are expressed at very low levels when compared with B. cereus. We found that activation of the PlcR regulon in B. anthracis by expression of a PlcR-PapR fusion protein does not alter sporulation in strains containing the virulence plasmid pXO1 and thereby the global regulator AtxA. Using comparative 2D gel electrophoresis, we showed that activation of the PlcR regulon in B. anthracis leads to upregulation of many proteins found in the secretome of B. cereus, including phospholipases and proteases, such as the putative protease BA1995. Transcriptional analysis demonstrated expression of BA1995 to be dependent on PlcR-PapR, even though the putative PlcR recognition site of the BA1995 gene does not exactly match the PlcR consensus sequence, explaining why this protein had escaped recognition as belonging to the PlcR regulon. Additionally, while transcription of major PlcR-dependent haemolysins, sphingomyelinase and anthrolysin O is enhanced in response to PlcR activation in B. anthracis, only anthrolysin O contributes significantly to lysis of human erythrocytes. In contrast, the toxicity of bacterial culture supernatants from a PlcR-positive strain towards murine macrophages occurred independently of anthrolysin O expression in vitro and in vivo.


Subject(s)
Bacillus anthracis/genetics , Bacterial Proteins/metabolism , Regulon , Trans-Activators/metabolism , Amino Acid Sequence , Animals , Anthrax/microbiology , Bacillus anthracis/pathogenicity , Bacillus cereus/genetics , Bacillus cereus/metabolism , Bacterial Proteins/genetics , Gene Expression Regulation, Bacterial , Humans , Macrophages/microbiology , Mice , Mice, Inbred DBA , Molecular Sequence Data , Mutagenesis , Plasmids , Recombinant Fusion Proteins/metabolism , Trans-Activators/genetics , Virulence
12.
Mol Pharm ; 7(3): 826-35, 2010 Jun 07.
Article in English | MEDLINE | ID: mdl-20230025

ABSTRACT

Toll-like receptor (TLR) agonists induce potent innate immune responses and can be used in the development of novel vaccine adjuvants. However, access to TLRs can be challenging as exemplified by TLR 7, which is located intracellularly in endosomal compartments. To increase recognition and subsequent stimulatory effects of TLR 7, imiquimod was encapsulated in acetalated dextran (Ac-DEX) microparticles. Ac-DEX, a water-insoluble and biocompatible polymer, is relatively stable at pH 7.4, but degrades rapidly under acidic conditions, such as those found in lysosomal vesicles. To determine the immunostimulatory capacity of encapsulated imiquimod, we compared the efficacy of free versus encapsulated imiquimod in activating RAW 264.7 macrophages, MH-S macrophages, and bone marrow derived dendritic cells. Encapsulated imiquimod significantly increased IL-1 beta, IL-6, and TNF-alpha cytokine expression in macrophages relative to the free drug. Furthermore, significant increases were observed in classic macrophage activation markers (iNOS, PD1-L1, and NO) after treatment with encapsulated imiquimod over the free drug. Also, bone marrow derived dendritic cells produced significantly higher levels of IL-1 beta, IL-6, IL-12p70, and MIP-1 alpha as compared to their counterparts receiving free imiquimod. These results suggest that encapsulation of TLR ligands within Ac-DEX microparticles results in increased immunostimulation and potentially better protection from disease when used in conjunction with vaccine formulations.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Dextrans/chemistry , Nanoparticles/chemistry , Adjuvants, Immunologic/chemistry , Aminoquinolines/administration & dosage , Aminoquinolines/chemistry , Animals , Cell Line , Cells, Cultured , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Imiquimod , Interleukin-12/metabolism , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Macrophage Activation/drug effects , Mice , Microscopy, Electron, Scanning , Nanoparticles/ultrastructure , Polymerase Chain Reaction
13.
J Leukoc Biol ; 82(6): 1531-41, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17761953

ABSTRACT

gp49B, an Ig-like receptor, negatively regulates the activity of mast cells and neutrophils through cytoplasmic immunoreceptor tyrosine-based inhibition motifs. To characterize the role of gp49B further in vivo, gp49B-deficient mice were tested in two allergic models. Responses to ragweed (RW) challenge in the lung and conjunctiva were assessed in models of allergic inflammation and during an infection with parasitic larvae of the nematode Ascaris suum. Infiltration by inflammatory cells into the lung during allergic responses was under negative control of the inhibitory receptor gp49B. Furthermore, an increase in conjunctival inflammation with a predominance of eosinophils, neutrophils, and degranulated mast cells was observed in RW-sensitized, gp49B-deficient mice, which had been challenged in the eye, as compared with C57BL/6 wild-type (WT) controls. Finally, an increase in allergic inflammation in the lungs of A. suum-infected, RW-sensitized mice was observed upon RW challenge, as compared with C57BL/6 WT controls. The observed influx of eosinophils into mucus membranes is characteristic of allergic asthma and allergic conjunctivitis and may contribute to airway hyper-responsiveness, airway remodeling, and mucus production. Expression of gp49B was detected on peripheral eosinophils of control mice and on eosinophils from lungs of mice treated with RW, suggesting a role for gp49B on eosinophils in dampening allergic inflammatory responses.


Subject(s)
Eosinophils/immunology , Hypersensitivity/immunology , Inflammation/immunology , Membrane Glycoproteins/immunology , Receptors, Immunologic/immunology , Allergens , Ambrosia , Animals , Ascaris suum/immunology , Ascaris suum/physiology , Bronchial Provocation Tests , Cell Degranulation , Cell Separation , Conjunctivitis, Allergic/immunology , Cytokines/metabolism , Eosinophilia/immunology , Flow Cytometry , Hypersensitivity/parasitology , Immunoglobulin Isotypes/blood , Inflammation/parasitology , Lung/immunology , Lung/parasitology , Lung/pathology , Mast Cells/cytology , Mast Cells/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Parasites/immunology , Parasites/physiology
14.
Curr Opin Allergy Clin Immunol ; 7(5): 424-8, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17873583

ABSTRACT

PURPOSE OF REVIEW: The purpose of this article is to summarize the clinical presentations associated with the classification of ocular allergy. This article also serves to summarize recent findings of pathophysiological mechanisms associated with ocular allergy and to highlight recently improved diagnostic methods for ocular allergic inflammation. RECENT FINDINGS: The term allergic conjunctivitis may not sufficiently describe all forms of allergic eye disease, thus a new classification system is desirable, preferably derived from the varied pathophysiological mechanisms operating in the different forms of ocular allergy. Recent published material has further characterized the roles that inflammatory and structural cells have in ocular allergic inflammation. Improved diagnostic methods have also been developed to assess the underlying causes of ocular allergy. SUMMARY: The underlying immune responses of ocular allergies are complex, indicating the critical need to understand the pathophysiology behind these diseases. Extensive research over the past several years has provided valuable insight into understanding the pathophysiology associated with the different forms of allergic conjunctivitis. Further clarification of the mechanisms associated with different forms of ocular allergy is essential for improved methods of classification, diagnosis, and treatment.


Subject(s)
Conjunctivitis, Allergic , Animals , Antigen-Presenting Cells/immunology , Conjunctivitis, Allergic/diagnosis , Conjunctivitis, Allergic/immunology , Conjunctivitis, Allergic/physiopathology , Cytokines/immunology , Cytokines/metabolism , Eye/immunology , Humans , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Keratoconjunctivitis/immunology , Mast Cells/immunology
15.
Int J Genomics ; 2017: 6489383, 2017.
Article in English | MEDLINE | ID: mdl-28791299

ABSTRACT

Increasing evidence that microRNAs (miRNAs) play important roles in the immune response against infectious agents suggests that miRNA might be exploitable as signatures of exposure to specific infectious agents. In order to identify potential early miRNA biomarkers of bacterial infections, human peripheral blood mononuclear cells (hPBMCs) were exposed to two select agents, Burkholderia pseudomallei K96243 and Francisella tularensis SHU S4, as well as to the nonpathogenic control Escherichia coli DH5α. RNA samples were harvested at three early time points, 30, 60, and 120 minutes postexposure, then sequenced. RNAseq analyses identified 87 miRNAs to be differentially expressed (DE) in a linear fashion. Of these, 31 miRNAs were tested using the miScript miRNA qPCR assay. Through RNAseq identification and qPCR validation, we identified differentially expressed miRNA species that may be involved in the early response to bacterial infections. Based upon its upregulation at early time points postexposure in two different individuals, hsa-mir-30c-5p is a miRNA species that could be studied further as a potential biomarker for exposure to these gram-negative intracellular pathogens. Gene ontology functional analyses demonstrated that programmed cell death is the first ranking biological process associated with miRNAs that are upregulated in F. tularensis-exposed hPBMCs.

16.
Adv Healthc Mater ; 5(20): 2617-2627, 2016 10.
Article in English | MEDLINE | ID: mdl-27594343

ABSTRACT

Subunit formulations are regarded as the safest type of vaccine, but they often contain a protein-based antigen that can result in significant challenges, such as preserving antigenicity during formulation and administration. Many studies have demonstrated that encapsulation of protein antigens in polymeric microparticles (MPs) via emulsion techniques results in total IgG antibody titers comparable to alum formulations, however, the antibodies themselves are non-neutralizing. To address this issue, a coaxial electrohydrodynamic spraying (electrospray) technique is used to formulate a microparticulate-based subunit anthrax vaccine under conditions that minimize recombinant protective antigen (rPA) exposure to harsh solvents and high shear stress. rPA and the adjuvant resiquimod are encapsulated either in separate or the same acetalated dextran MPs. Using a murine model, the electrospray formulations lead to higher IgG2a subtype titers as well as comparable total IgG antibody titers and toxin neutralization relative to the FDA-approved vaccine (BioThrax). BioThrax provides no protection against a lethal inhalational challenge of the highly virulent Ames Bacillus anthracis anthrax strain, whereas 50% of the mice vaccinated with separately encapsulated electrospray MPs survive. Overall, this study demonstrates the potential use of electrospray for encapsulating protein antigens in polymeric MPs.


Subject(s)
Antibodies, Neutralizing/immunology , Bacillus anthracis/immunology , Bacterial Toxins/immunology , Dextrans/chemistry , Dextrans/immunology , Vaccines/chemistry , Vaccines/immunology , Animals , Anthrax/immunology , Anthrax Vaccines/immunology , Antigens, Bacterial/immunology , Chemistry, Pharmaceutical/methods , Female , Immunoglobulin G/immunology , Mice , Mice, Inbred BALB C , Polymers/chemistry
17.
Invest Ophthalmol Vis Sci ; 46(8): 2772-80, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16043850

ABSTRACT

PURPOSE: To assess alterations in allergic ocular responses to nonparasite antigens in an experimental system in which mice were skewed toward a Th2 cytokine profile by helminth infection. METHODS: Mice were inoculated with Ascaris suum (A. suum) eggs concurrent with ragweed (RW) sensitization (RW/acute) or by repeated inoculation before RW sensitization (RW/chronic). Control subjects were divided into RW, A. suum, and sham-sensitized groups. Animals were RW-challenged in the eye and examined for changes in ocular responses, inflammatory cell infiltrates, and in vitro assessment of cytokines after antigen restimulation. In subsequent experiments, CD4(+)/CD25+ T regulatory and CD4(+)/CD25- control T cells were adoptively transferred into mice before ocular challenge. RESULTS: RW sensitization and challenge increased ocular symptoms and eosinophil infiltration into the conjunctiva over PBS control eyes. Acute A. suum infection significantly increased RW-induced clinical symptoms and eosinophil infiltrates in the conjunctiva (P = 0.0001) and resulted in the development of anterior uveitis. In contrast, RW/chronic infection provided protection from allergic responses to RW with significantly fewer eosinophils in the eye and reduced eotaxin levels. Transfer of CD4(+)/CD25+ T cells from RW/chronic mice into RW/acute animals also decreased disease intensity, suggesting that T regulatory cells may contribute to protection from allergic eye disease. CONCLUSIONS: The current studies suggest acute parasitic infections exacerbate allergic symptoms, whereas chronic infections offer protection and provide possible explanations for the role of parasitic infection in susceptibility and resistance to nonparasite allergens.


Subject(s)
Ascariasis/immunology , Ascaris suum/immunology , Blepharitis/immunology , Conjunctivitis, Allergic/immunology , Hypersensitivity/immunology , Uveitis, Anterior/immunology , Acute Disease , Adoptive Transfer , Allergens/immunology , Ambrosia/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , Chemokine CCL11 , Chemokines, CC/blood , Chronic Disease , Enzyme-Linked Immunosorbent Assay , Eosinophilia/immunology , Eosinophils/immunology , Immunoglobulin E/blood , Interleukin-6/biosynthesis , Mast Cells/immunology , Mice , Mice, Inbred A , Mice, Inbred BALB C , Mice, Knockout , Th2 Cells/immunology
18.
Curr Opin Allergy Clin Immunol ; 3(5): 375-9, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14501438

ABSTRACT

PURPOSE OF REVIEW: This review focuses on reports on animal models of ocular allergy published within the past year. A number of animal models are currently being used to clarify the pathophysiology of ocular allergy and to improve the therapeutic interventions for this disease. RECENT FINDINGS: Published literature examined the role of cytokines and other effector molecules which drive the immunopathology of ocular allergies in several animal models. Animal models were also used to compare the safety and efficacy of currently available drugs, and were utilized in initial trials of novel therapeutic agents. Novel therapeutic options being studied include DNA immunizations and recombinant peptides that block enzymes involved in the inflammatory processes. SUMMARY: Several animal models are currently being used in the study of ocular allergy. These include different strains within the mouse, rat, guinea pig, rabbit and dog species. Continuing investigations are needed to elucidate the complex molecular and cellular processes involved in the pathogenesis of ocular allergies. A better understanding of the interplay of effector cells, cytokines, adhesion molecules and a number of other inflammatory mediators will broaden our knowledge of the pathophysiology of ocular allergy and allow improved therapeutic options for this disease.


Subject(s)
Eye Diseases/immunology , Hypersensitivity/complications , Animals , Dogs , Eye Diseases/drug therapy , Eye Diseases/physiopathology , Guinea Pigs , Humans , Hypersensitivity/drug therapy , Hypersensitivity/physiopathology , Mice , Models, Animal , Rabbits , Rats
19.
Methods Mol Biol ; 1121: 269-78, 2014.
Article in English | MEDLINE | ID: mdl-24510831

ABSTRACT

Vaccines have evolved for hundreds of years, but all utilize the premise that safely pre-exposing the host to some component of a pathogen allows for enhanced immune recognition, and potential protection from disease, upon encountering the pathogen at a later date. Early vaccination strategies used inactivated or attenuated vaccines, many of which contained toxins and other components that resulted in reactogenicity or risk of reversion to virulence. DNA vaccines supplant many of the issues associated with inactivated or attenuated vaccines, but these vaccines tend to provide weak immunological responses, particularly in primates. DNA Electroporation may prove to be the "missing link" in the evolution of DNA vaccines allowing for enhanced immune responses from DNA vaccination in humans thereby resulting in protection from disease post-pathogen exposure.


Subject(s)
Electroporation/methods , Vaccines, DNA/metabolism , Animals , DNA/immunology , DNA/metabolism , Epidermis , Humans , Immunization , Vaccines, DNA/immunology
20.
Methods Mol Biol ; 1121: 325-36, 2014.
Article in English | MEDLINE | ID: mdl-24510836

ABSTRACT

Effective multi-agent/multivalent vaccines that confer protection against more than one disease are highly desirable to the patient and to health-care professionals. Electroporation of DNA vaccines, whereby tissues injected with DNA are subjected to localized electrical currents, is an ideal platform technology that achieves protective immune responses to multivalent vaccination. Here, we describe an electroporation-based immunization technique capable of administering a cocktail of DNA vaccinations in vivo. Immune response measurements, including protection from pathogen challenge and induction of antigen-specific antibody responses and cell-mediated immune responses, are also discussed.


Subject(s)
Electroporation/methods , Vaccines, DNA/immunology , Vaccines, DNA/metabolism , Animals , Electrodes , Electroporation/instrumentation , Female , Mice , Plasmids/genetics
SELECTION OF CITATIONS
SEARCH DETAIL