Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Appl Microbiol Biotechnol ; 101(13): 5313-5324, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28405704

ABSTRACT

Increasing demand for the low-cost production of valuable proteins has stimulated development of novel expression systems. Many challenges faced by existing technology may be overcome by using unicellular microalgae as an expression platform due to their ability to be cultivated rapidly, inexpensively, and in large scale. Diatoms are a particularly productive type of unicellular algae showing promise as production organisms. Here, we report the development of an expression system in the diatom Thalassiosira pseudonana by expressing the protective IbpA DR2 antigen from Histophilus somni for the production of a vaccine against bovine respiratory disease. The utilization of diatoms with their typically silicified cell walls permitted development of silicon-responsive transcription elements to induce protein expression. Specifically, we demonstrate that transcription elements from the silicon transporter gene SIT1 are sufficient to drive high levels of IbpA DR2 expression during silicon limitation and growth arrest. These culture conditions eliminate the flux of cellular resources into cell division processes, yet do not limit protein expression. In addition to improving protein expression levels by molecular manipulations, yield was dramatically increased through cultivation enhancement including elevated light and CO2 supplementation. We substantially increased recombinant protein production over starting levels to 1.2% of the total sodium dodecyl sulfate-extractable protein in T. pseudonana, which was sufficient to conduct preliminary immunization trials in mice. Mice exposed to 5 µg of diatom-expressed DR2 in whole or sonicated cells (without protein purification) exhibited a modest immune response without the addition of adjuvant.


Subject(s)
Antigens, Bacterial/biosynthesis , Cattle Diseases/prevention & control , Diatoms/genetics , Pasteurellaceae Infections/veterinary , Pasteurellaceae/genetics , Animals , Antibodies, Bacterial , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Bacterial Vaccines/genetics , Bacterial Vaccines/immunology , Carbon Dioxide/metabolism , Carbon Dioxide/pharmacology , Cattle , Cattle Diseases/microbiology , Diatoms/drug effects , Diatoms/growth & development , Diatoms/metabolism , Light , Mice , Pasteurellaceae Infections/immunology , Pasteurellaceae Infections/prevention & control , Recombinant Proteins/administration & dosage , Recombinant Proteins/biosynthesis , Recombinant Proteins/immunology , Recombinant Proteins/isolation & purification , Respiratory Tract Infections/immunology , Respiratory Tract Infections/microbiology , Respiratory Tract Infections/prevention & control , Respiratory Tract Infections/veterinary , Silicon/pharmacology , Transcription Factors/genetics , Transcription Factors/metabolism , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
2.
Vaccine ; 35(15): 1954-1963, 2017 04 04.
Article in English | MEDLINE | ID: mdl-28274639

ABSTRACT

Bovine respiratory syncytial virus (BRSV) and Histophilus somni synergize to cause respiratory disease in cattle. These pathogens cause enhanced disease during dual-infection and an IgE response to antigens of H. somni in dual-infected but not singly infected calves. Vaccines containing whole inactivated BRSV or H. somni have been associated with IgE responses A vaccine strategy that avoids stimulation of IgE antibodies would provide superior protection from dual infection. We hypothesized that a subunit vaccine consisting of the nucleoprotein (NP) from BRSV and the recombinant antigen IbpA DR2 (a surface antigen of H. somni with two toxic fic motifs) in Quil A adjuvant would elicit protection without disease enhancement. Three groups of calves were vaccinated twice with either: Formalin inactivated BRSV (FI) plus Somnivac®, NP & IbpA DR2 plus Quil A or Quil A alone, followed by BRSV and H. somni challenge. Clinical scores and antibody levels (to whole pathogens and to the subunits) were evaluated. Lungs were examined at necropsy on day 23 after infection. Clinical scores were significantly greatest for the FI & Somnivac® group and both clinical scores and lung pathology were lowest for the subunit group. All calves shed BRSV in nasal secretions. FI & Somnivac® induced IgE antibodies to H. somni and BRSV, but not to NP or DR2. The subunit vaccine did not induce an IgE antibody response to IbpA DR2 antigen and induced little IgE to H. somni. It did not induce an IgG antibody response to BRSV and H. somni, but stimulated production of IgG antibodies against the subunits. In summary, the subunit vaccine, consisting of the BRSV NP and H. somni IbpA DR2 in Quil A, protected against severe clinical signs and decreased lung pathology but did not prevent viral shedding. Importantly it prevented synergistic disease expression in response to dual infection.


Subject(s)
Bacterial Vaccines/immunology , Pasteurellaceae Infections/veterinary , Pasteurellaceae/immunology , Respiratory Syncytial Virus Infections/veterinary , Respiratory Syncytial Viruses/immunology , Viral Vaccines/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Antibody Formation , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/genetics , Cattle , Lung/pathology , Pasteurellaceae Infections/pathology , Pasteurellaceae Infections/prevention & control , Quillaja Saponins/administration & dosage , Respiratory Syncytial Virus Infections/prevention & control , Severity of Illness Index , Treatment Outcome , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/genetics
3.
J Allergy Clin Immunol ; 136(3): 747-756.e4, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25930197

ABSTRACT

BACKGROUND: Single nucleotide polymorphisms in the human gene for the receptor for advanced glycation end-products (RAGE) are associated with an increased incidence of asthma. RAGE is highly expressed in the lung and has been reported to play a vital role in the pathogenesis of murine models of asthma/allergic airway inflammation (AAI) by promoting expression of the type 2 cytokines IL-5 and IL-13. IL-5 and IL-13 are prominently secreted by group 2 innate lymphoid cells (ILC2s), which are stimulated by the proallergic cytokine IL-33. OBJECTIVE: We sought to test the hypothesis that pulmonary RAGE is necessary for allergen-induced ILC2 accumulation in the lung. METHODS: AAI was induced in wild-type and RAGE knockout mice by using IL-33, house dust mite extract, or Alternaria alternata extract. RAGE's lung-specific role in type 2 responses was explored with bone marrow chimeras and induction of gastrointestinal type 2 immune responses. RESULTS: RAGE was found to drive AAI by promoting IL-33 expression in response to allergen and by coordinating the inflammatory response downstream of IL-33. Absence of RAGE impedes pulmonary accumulation of ILC2s in models of AAI. Bone marrow chimera studies suggest that pulmonary parenchymal, but not hematopoietic, RAGE has a central role in promoting AAI. In contrast to the lung, the absence of RAGE does not affect IL-33-induced ILC2 influx in the spleen, type 2 cytokine production in the peritoneum, or mucus hypersecretion in the gastrointestinal tract. CONCLUSIONS: For the first time, this study demonstrates that a parenchymal factor, RAGE, mediates lung-specific accumulation of ILC2s.


Subject(s)
Asthma/immunology , Immunity, Innate , Interleukin-33/immunology , Lung/immunology , Lymphocytes/immunology , Receptor for Advanced Glycation End Products/immunology , Allergens/administration & dosage , Allergens/immunology , Alternaria/chemistry , Animals , Antigens, Dermatophagoides/administration & dosage , Antigens, Dermatophagoides/immunology , Asthma/chemically induced , Asthma/genetics , Asthma/pathology , Bone Marrow/immunology , Bone Marrow/pathology , Cell Proliferation , Gastrointestinal Tract/immunology , Gastrointestinal Tract/pathology , Gene Expression Regulation , Interleukin-13/genetics , Interleukin-13/immunology , Interleukin-33/genetics , Interleukin-5/genetics , Interleukin-5/immunology , Lung/pathology , Lymphocytes/pathology , Mice , Organ Specificity , Peritoneum/immunology , Peritoneum/pathology , Pyroglyphidae/chemistry , Receptor for Advanced Glycation End Products/genetics , Signal Transduction , Spleen/immunology , Spleen/pathology , Transplantation Chimera
4.
J Leukoc Biol ; 98(2): 143-52, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25743626

ABSTRACT

IPF is a progressive lung disorder characterized by fibroblast proliferation and myofibroblast differentiation. Although neutrophil accumulation within IPF lungs has been negatively correlated with outcomes, the role played by neutrophils in lung fibrosis remains poorly understood. We have demonstrated previously that NE promotes lung cancer cell proliferation and hypothesized that it may have a similar effect on fibroblasts. In the current study, we show that NE(-/-) mice are protected from asbestos-induced lung fibrosis. NE(-/-) mice displayed reduced fibroblast and myofibroblast content when compared with controls. NE directly both lung fibroblast proliferation and myofibroblast differentiation in vitro, as evidenced by proliferation assays, collagen gel contractility assays, and αSMA induction. Furthermore, αSMA induction occurs in a TGF-ß-independent fashion. Treatment of asbestos-recipient mice with ONO-5046, a synthetic NE antagonist, reduced hydroxyproline content. Thus, the current study points to a key role for neutrophils and NE in the progression of lung fibrosis. Lastly, the study lends rationale to use of NE-inhibitory approaches as a novel therapeutic strategy for patients with lung fibrosis.


Subject(s)
Leukocyte Elastase/metabolism , Lung/enzymology , Myofibroblasts/enzymology , Neutrophils/enzymology , Pulmonary Emphysema/enzymology , Pulmonary Fibrosis/enzymology , Animals , Cell Differentiation , Cell Proliferation , Disease Models, Animal , Gene Expression Regulation , Humans , Insulin Receptor Substrate Proteins/genetics , Insulin Receptor Substrate Proteins/immunology , Leukocyte Elastase/genetics , Leukocyte Elastase/immunology , Lung/immunology , Lung/pathology , Mice , Myofibroblasts/immunology , Myofibroblasts/pathology , Neutrophils/immunology , Neutrophils/pathology , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/immunology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology , Pulmonary Emphysema/genetics , Pulmonary Emphysema/immunology , Pulmonary Emphysema/pathology , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/immunology , Pulmonary Fibrosis/pathology , Signal Transduction
5.
PLoS One ; 9(3): e88259, 2014.
Article in English | MEDLINE | ID: mdl-24642901

ABSTRACT

Elucidating the sites and mechanisms of sRAGE action in the healthy state is vital to better understand the biological importance of the receptor for advanced glycation end products (RAGE). Previous studies in animal models of disease have demonstrated that exogenous sRAGE has an anti-inflammatory effect, which has been reasoned to arise from sequestration of pro-inflammatory ligands away from membrane-bound RAGE isoforms. We show here that sRAGE exhibits in vitro binding with high affinity and reversibly to extracellular matrix components collagen I, collagen IV, and laminin. Soluble RAGE administered intratracheally, intravenously, or intraperitoneally, does not distribute in a specific fashion to any healthy mouse tissue, suggesting against the existence of accessible sRAGE sinks and receptors in the healthy mouse. Intratracheal administration is the only effective means of delivering exogenous sRAGE to the lung, the organ in which RAGE is most highly expressed; clearance of sRAGE from lung does not differ appreciably from that of albumin.


Subject(s)
Collagen Type IV/metabolism , Collagen Type I/metabolism , Laminin/metabolism , Receptors, Immunologic/metabolism , Administration, Inhalation , Animals , Biological Availability , Fibronectins/metabolism , Humans , Injections, Intraperitoneal , Injections, Intravenous , Kinetics , Lung/chemistry , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Protein Binding , Receptor for Advanced Glycation End Products , Receptors, Immunologic/administration & dosage , Receptors, Immunologic/isolation & purification , Solubility
6.
Am J Pathol ; 181(4): 1215-25, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22889845

ABSTRACT

The receptor for advanced glycation end products (RAGE) is a multiligand receptor that has been shown to contribute to the pathogenesis of diabetes, atherosclerosis, and neurodegeneration. However, its role in asthma and allergic airway disease is largely unknown. These studies use a house dust mite (HDM) mouse model of asthma/allergic airway disease. Respiratory mechanics were assessed and compared between wild-type and RAGE knockout mice. Bronchovascular architecture was assessed with quantitative scoring, and expression of RAGE, immunoglobulins, and relevant cytokines was assessed by standard protein detection methods and/or quantitative RT-PCR. The absence of RAGE abolishes most assessed measures of pathology, including airway hypersensitivity (resistance, tissue damping, and elastance), eosinophilic inflammation, and airway remodeling. IL-4 secretion, isotype class switching, and antigen recognition are intact in the absence of RAGE. In contrast, normal increases in IL-5, IL-13, eotaxin, and eotaxin-2 production are abrogated in the RAGE knockouts. IL-17 indicates complex regulation, with elevated baseline expression in RAGE knockouts, but no induction in response to allergen. Treatment of WT mice with an inhibitor of RAGE markedly reduces inflammation in the HDM model, suggesting that RAGE inhibition may serve as a promising therapeutic strategy. Finally, the results in the HDM model are recapitulated in an ovalbumin model of asthma, suggesting that RAGE plays a role in asthma irrespective of the identity of the allergens involved.


Subject(s)
Asthma/etiology , Asthma/metabolism , Receptors, Immunologic/metabolism , Animals , Asthma/pathology , Asthma/physiopathology , Bronchial Hyperreactivity/parasitology , Bronchial Hyperreactivity/pathology , Bronchial Hyperreactivity/physiopathology , Chemokine CCL24 , Eosinophilia/parasitology , Eosinophilia/pathology , Eosinophilia/physiopathology , Immunoglobulin G/immunology , Interleukin-13/biosynthesis , Interleukin-4/biosynthesis , Interleukin-4/metabolism , Interleukin-5/biosynthesis , Lung/metabolism , Lung/pathology , Lung/physiopathology , Male , Mice , Mice, Inbred C57BL , Ovalbumin , Protein Transport , Pyroglyphidae/physiology , Receptor for Advanced Glycation End Products
SELECTION OF CITATIONS
SEARCH DETAIL
...