Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Appl Microbiol Biotechnol ; 101(13): 5313-5324, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28405704

RESUMEN

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.


Asunto(s)
Antígenos Bacterianos/biosíntesis , Enfermedades de los Bovinos/prevención & control , Diatomeas/genética , Infecciones por Pasteurellaceae/veterinaria , Pasteurellaceae/genética , Animales , Anticuerpos Antibacterianos , Antígenos Bacterianos/genética , Antígenos Bacterianos/inmunología , Vacunas Bacterianas/genética , Vacunas Bacterianas/inmunología , Dióxido de Carbono/metabolismo , Dióxido de Carbono/farmacología , Bovinos , Enfermedades de los Bovinos/microbiología , Diatomeas/efectos de los fármacos , Diatomeas/crecimiento & desarrollo , Diatomeas/metabolismo , Luz , Ratones , Infecciones por Pasteurellaceae/inmunología , Infecciones por Pasteurellaceae/prevención & control , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/aislamiento & purificación , Infecciones del Sistema Respiratorio/inmunología , Infecciones del Sistema Respiratorio/microbiología , Infecciones del Sistema Respiratorio/prevención & control , Infecciones del Sistema Respiratorio/veterinaria , Silicio/farmacología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología
2.
J Allergy Clin Immunol ; 136(3): 747-756.e4, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25930197

RESUMEN

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.


Asunto(s)
Asma/inmunología , Inmunidad Innata , Interleucina-33/inmunología , Pulmón/inmunología , Linfocitos/inmunología , Receptor para Productos Finales de Glicación Avanzada/inmunología , Alérgenos/administración & dosificación , Alérgenos/inmunología , Alternaria/química , Animales , Antígenos Dermatofagoides/administración & dosificación , Antígenos Dermatofagoides/inmunología , Asma/inducido químicamente , Asma/genética , Asma/patología , Médula Ósea/inmunología , Médula Ósea/patología , Proliferación Celular , Tracto Gastrointestinal/inmunología , Tracto Gastrointestinal/patología , Regulación de la Expresión Génica , Interleucina-13/genética , Interleucina-13/inmunología , Interleucina-33/genética , Interleucina-5/genética , Interleucina-5/inmunología , Pulmón/patología , Linfocitos/patología , Ratones , Especificidad de Órganos , Peritoneo/inmunología , Peritoneo/patología , Pyroglyphidae/química , Receptor para Productos Finales de Glicación Avanzada/genética , Transducción de Señal , Bazo/inmunología , Bazo/patología , Quimera por Trasplante
3.
Am J Pathol ; 181(4): 1215-25, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22889845

RESUMEN

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.


Asunto(s)
Asma/etiología , Asma/metabolismo , Receptores Inmunológicos/metabolismo , Animales , Asma/patología , Asma/fisiopatología , Hiperreactividad Bronquial/parasitología , Hiperreactividad Bronquial/patología , Hiperreactividad Bronquial/fisiopatología , Quimiocina CCL24 , Eosinofilia/parasitología , Eosinofilia/patología , Eosinofilia/fisiopatología , Inmunoglobulina G/inmunología , Interleucina-13/biosíntesis , Interleucina-4/biosíntesis , Interleucina-4/metabolismo , Interleucina-5/biosíntesis , Pulmón/metabolismo , Pulmón/patología , Pulmón/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Ovalbúmina , Transporte de Proteínas , Pyroglyphidae/fisiología , Receptor para Productos Finales de Glicación Avanzada
4.
Vaccine ; 35(15): 1954-1963, 2017 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-28274639

RESUMEN

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.


Asunto(s)
Vacunas Bacterianas/inmunología , Infecciones por Pasteurellaceae/veterinaria , Pasteurellaceae/inmunología , Infecciones por Virus Sincitial Respiratorio/veterinaria , Virus Sincitiales Respiratorios/inmunología , Vacunas Virales/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Animales , Formación de Anticuerpos , Vacunas Bacterianas/administración & dosificación , Vacunas Bacterianas/genética , Bovinos , Pulmón/patología , Infecciones por Pasteurellaceae/patología , Infecciones por Pasteurellaceae/prevención & control , Saponinas de Quillaja/administración & dosificación , Infecciones por Virus Sincitial Respiratorio/prevención & control , Índice de Severidad de la Enfermedad , Resultado del Tratamiento , Vacunas de Subunidad/administración & dosificación , Vacunas de Subunidad/genética , Vacunas de Subunidad/inmunología , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Vacunas Virales/administración & dosificación , Vacunas Virales/genética
5.
J Leukoc Biol ; 98(2): 143-52, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25743626

RESUMEN

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.


Asunto(s)
Elastasa de Leucocito/metabolismo , Pulmón/enzimología , Miofibroblastos/enzimología , Neutrófilos/enzimología , Enfisema Pulmonar/enzimología , Fibrosis Pulmonar/enzimología , Animales , Diferenciación Celular , Proliferación Celular , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Humanos , Proteínas Sustrato del Receptor de Insulina/genética , Proteínas Sustrato del Receptor de Insulina/inmunología , Elastasa de Leucocito/genética , Elastasa de Leucocito/inmunología , Pulmón/inmunología , Pulmón/patología , Ratones , Miofibroblastos/inmunología , Miofibroblastos/patología , Neutrófilos/inmunología , Neutrófilos/patología , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/inmunología , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/inmunología , Enfisema Pulmonar/genética , Enfisema Pulmonar/inmunología , Enfisema Pulmonar/patología , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/inmunología , Fibrosis Pulmonar/patología , Transducción de Señal
6.
PLoS One ; 9(3): e88259, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24642901

RESUMEN

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.


Asunto(s)
Colágeno Tipo IV/metabolismo , Colágeno Tipo I/metabolismo , Laminina/metabolismo , Receptores Inmunológicos/metabolismo , Administración por Inhalación , Animales , Disponibilidad Biológica , Fibronectinas/metabolismo , Humanos , Inyecciones Intraperitoneales , Inyecciones Intravenosas , Cinética , Pulmón/química , Pulmón/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Unión Proteica , Receptor para Productos Finales de Glicación Avanzada , Receptores Inmunológicos/administración & dosificación , Receptores Inmunológicos/aislamiento & purificación , Solubilidad
7.
Int J Clin Exp Pathol ; 4(4): 349-55, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21577320

RESUMEN

AIM: The purpose of this study was to develop an improved method for collagen and protein assessment of fibrotic lungs while decreasing animal use. METHODS: 8-10 week old, male C57BL/6 mice were given a single intratracheal instillation of crocidolite asbestos or control titanium dioxide. Lungs were collected on day 14 and dried as whole lung, or homogenized in CHAPS buffer, for hydroxyproline analysis. Insoluble and salt-soluble collagen content was also determined in lung homogenates using a modified Sirius red colorimetric 96-well plate assay. RESULTS: The hydroxyproline assay showed significant increases in collagen content in the lungs of asbestos-treated mice. Identical results were present between collagen content determined on dried whole lung or whole lung homogenates. The Sirius red plate assay showed a significant increase in collagen content in lung homogenates however, this assay grossly over-estimated the total amount of collagen and underestimated changes between control and fibrotic lungs, conclusions: The proposed method provides accurate quantification of collagen content in whole lungs and additional homogenate samples for biochemical analysis from a single animal. The Sirius-red colorimetric plate assay provides a complementary method for determination of the relative changes in lung collagen but the values tend to overestimate absolute values obtained by the gold standard hydroxyproline assay and underestimate the overall fibrotic injury.


Asunto(s)
Asbestosis/metabolismo , Colágeno/metabolismo , Colorimetría , Pulmón/metabolismo , Fibrosis Pulmonar/metabolismo , Análisis de Varianza , Animales , Asbesto Crocidolita , Asbestosis/etiología , Asbestosis/patología , Compuestos Azo , Biomarcadores/metabolismo , Colorimetría/normas , Colorantes , Modelos Animales de Enfermedad , Hidroxiprolina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/patología , Reproducibilidad de los Resultados , Índice de Severidad de la Enfermedad , Espectrofotometría , Regulación hacia Arriba
8.
Int J Clin Exp Pathol ; 4(3): 241-54, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21487520

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a progressive disease with poor survival. The identification of therapeutic targets is essential to improving outcomes. Previous studies found that expression of the receptor for advanced glycation end products (RAGE) in the lung is significantly decreased in human IPF lungs and in two animal models of pulmonary fibrosis. In addition, RAGE-null mice spontaneously develop pulmonary fibrosis with age and more severe fibrosis when challenged with asbestos. In contrast to the findings that the lack of RAGE enhanced pulmonary fibrosis, He et al. found that RAGE null mice were protected from bleomycin-induced fibrosis and suggested the effect was due to a lack of HMGB1 induced RAGE signaling. The current study further tests this hypothesis by blocking RAGE signaling via administration of soluble RAGE, a decoy receptor, to determine if this will also protect against pulmonary fibrosis. Wild-type, RAGE(+/-), and RAGE(-/-) mice were treated with bleomycin and assessed for fibrosis. Wild-type mice were also treated with exogenous soluble RAGE or vehicle control. In addition, in vitro studies with primary alveolar epithelial cells from wild-type and RAGE null mice were used to investigate the effect of RAGE on cell viability and migration in response to injury. A lack of RAGE was found to be protective against bleomycin injury in both in vivo and in vitro studies. However, soluble RAGE administration was unable to ameliorate fibrosis. This study confirms paradoxical responses to two different models of pulmonary fibrosis and suggests a further role for RAGE in cellular migration.


Asunto(s)
Fibrosis Pulmonar/metabolismo , Receptores Inmunológicos/metabolismo , Análisis de Varianza , Animales , Bleomicina , Western Blotting , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fibrosis Pulmonar/inducido químicamente , Receptor para Productos Finales de Glicación Avanzada , Receptores Inmunológicos/genética , Transducción de Señal
9.
PLoS One ; 6(5): e20132, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21629785

RESUMEN

BACKGROUND: The receptor for advanced glycation end-products (RAGE) has been suggested to modulate lung injury in models of acute pulmonary inflammation. To study this further, model systems utilizing wild type and RAGE knockout (KO) mice were used to determine the role of RAGE signaling in lipopolysaccharide (LPS) and E. coli induced acute pulmonary inflammation. The effect of intraperitoneal (i.p.) and intratracheal (i.t.) administration of mouse soluble RAGE on E. coli injury was also investigated. METHODOLOGY/PRINCIPAL FINDINGS: C57BL/6 wild type and RAGE KO mice received an i.t. instillation of LPS, E. coli, or vehicle control. Some groups also received i.p. or i.t. administration of mouse soluble RAGE. After 24 hours, the role of RAGE expression on inflammation was assessed by comparing responses in wild type and RAGE KO. RAGE protein levels decreased in wild type lung homogenates after treatment with either LPS or bacteria. In addition, soluble RAGE and HMGB1 increased in the BALF after E. coli instillation. RAGE KO mice challenged with LPS had the same degree of inflammation as wild type mice. However, when challenged with E. coli, RAGE KO mice had significantly less inflammation when compared to wild type mice. Most cytokine levels were lower in the BALF of RAGE KO mice compared to wild type mice after E. coli injury, while only monocyte chemotactic protein-1, MCP-1, was lower after LPS challenge. Neither i.p. nor i.t. administration of mouse soluble RAGE attenuated the severity of E. coli injury in wild type mice. CONCLUSIONS/SIGNIFICANCE: Lack of RAGE in the lung does not protect against LPS induced acute pulmonary inflammation, but attenuates injury following live E. coli challenge. These findings suggest that RAGE mediates responses to E. coli-associated pathogen-associated molecular pattern molecules other than LPS or other bacterial specific signaling responses. Soluble RAGE treatment had no effect on inflammation.


Asunto(s)
Escherichia coli/patogenicidad , Pulmón/metabolismo , Pulmón/microbiología , Neumonía/metabolismo , Neumonía/microbiología , Receptores Inmunológicos/metabolismo , Animales , Western Blotting , Líquido del Lavado Bronquioalveolar/química , Células Cultivadas , Quimiocina CCL2/metabolismo , Quimiocina CCL3/metabolismo , Técnicas In Vitro , Interleucina-12/metabolismo , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Peroxidasa/genética , Peroxidasa/metabolismo , Neumonía/genética , Receptor para Productos Finales de Glicación Avanzada , Receptores Inmunológicos/genética , Factor de Necrosis Tumoral alfa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA