Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Exp Bot ; 74(21): 6860-6873, 2023 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-37696760

RESUMEN

MYZUS PERSICAE-INDUCED LIPASE1 (MPL1) encodes a lipase in Arabidopsis thaliana that is required for limiting infestation by the green peach aphid (GPA; Myzus persicae), an important phloem sap-consuming insect pest. Previously, we demonstrated that MPL1 expression was up-regulated in response to GPA infestation, and GPA fecundity was higher on the mpl1 mutant, compared with the wild-type (WT), and lower on 35S:MPL1 plants that constitutively expressed MPL1 from the 35S promoter. Here, we show that the MPL1 promoter is active in the phloem and expression of the MPL1 coding sequence from the phloem-specific SUC2 promoter in mpl1 is sufficient to restore resistance to GPA. The GPA infestation-associated up-regulation of MPL1 requires CYCLOPHILIN 20-3 (CYP20-3), which encodes a 12-oxo-phytodienoic acid (OPDA)-binding protein that is involved in OPDA signaling, and is required for limiting GPA infestation. OPDA promotes MPL1 expression to limit GPA fecundity, a process that requires CYP20-3 function. These results along with our observation that constitutive expression of MPL1 from the 35S promoter restores resistance to GPA in the cyp20-3 mutant, and MPL1 acts in a feedback loop to limit OPDA levels in GPA-infested plants, suggest that an interplay between MPL1, OPDA, and CYP20-3 contributes to resistance to GPA.


Asunto(s)
Áfidos , Proteínas de Arabidopsis , Arabidopsis , Animales , Arabidopsis/genética , Arabidopsis/metabolismo , Lipasa/genética , Lipasa/metabolismo , Áfidos/fisiología , Proteínas de Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Hidrolasas de Éster Carboxílico/metabolismo , Mutación , Enfermedades de las Plantas , Regulación de la Expresión Génica de las Plantas
2.
Int J Mol Sci ; 22(16)2021 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-34445253

RESUMEN

Bronchopulmonary dysplasia (BPD) is the most common complication of prematurity and a key contributor to the large health care burden associated with prematurity, longer hospital stays, higher hospital costs, and frequent re-hospitalizations of affected patients through the first year of life and increased resource utilization throughout childhood. This disease is associated with abnormal pulmonary function that may lead to BPD-associated pulmonary hypertension (PH), a major contributor to neonatal mortality and morbidity. In the absence of any definitive treatment options, this life-threatening disease is associated with high resource utilization during and after neonatal intensive care unit (NICU) stay. The goal of this study was to test the safety and efficacy of a small molecule derivative of chitin, AVR-48, as prophylactic therapy for preventing experimental BPD in a mouse model. Two doses of AVR-48 were delivered either intranasally (0.11 mg/kg), intraperitoneally (10 mg/kg), or intravenously (IV) (10 mg/kg) to newborn mouse pups on postnatal day (P)2 and P4. The outcomes were assessed by measuring total inflammatory cells in the broncho-alveolar lavage fluid (BALF), chord length, septal thickness, and radial alveolar counts of the alveoli, Fulton's Index (for PH), cell proliferation and cell death by immunostaining, and markers of inflammation by Western blotting and ELISA. The bioavailability and safety of the drug were assessed by pharmacokinetic and toxicity studies in both neonatal mice and rat pups (P3-P5). Following AVR-48 treatment, alveolar simplification was improved, as evident from chord length, septal thickness, and radial alveolar counts; total inflammatory cells were decreased in the BALF; Fulton's Index was decreased and lung inflammation and cell death were decreased, while angiogenesis and cell proliferation were increased. AVR-48 was found to be safe and the no-observed-adverse-effect level (NOAEL) in rat pups was determined to be 100 mg/kg when delivered via IV dosing with a 20-fold safety margin. With no reported toxicity and with a shorter half-life, AVR-48 is able to reverse the worsening cardiopulmonary phenotype of experimental BPD and BPD-PH, compared to controls, thus positioning it as a future drug candidate.


Asunto(s)
Displasia Broncopulmonar , Quitina , Hipertensión Pulmonar , Neovascularización Fisiológica/efectos de los fármacos , Alveolos Pulmonares , Animales , Animales Recién Nacidos , Displasia Broncopulmonar/tratamiento farmacológico , Displasia Broncopulmonar/metabolismo , Displasia Broncopulmonar/patología , Quitina/química , Quitina/farmacología , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Hipertensión Pulmonar/tratamiento farmacológico , Hipertensión Pulmonar/metabolismo , Hipertensión Pulmonar/patología , Ratones , Alveolos Pulmonares/metabolismo , Alveolos Pulmonares/patología , Ratas
3.
Mol Vis ; 27: 37-49, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33633438

RESUMEN

Purpose: Glaucoma is a neurodegenerative disease of the eye with an estimated prevalence of more than 111.8 million patients worldwide by 2040, with at least 6 to 8 million projected to become bilaterally blind. Clinically, the current method of slowing glaucomatous vision loss is to reduce intraocular pressure (IOP). In this manuscript, we describe the in vitro cytoprotective and in vivo long lasting IOP-lowering activity of the poly D, L-lactic-co-glycolic acid (PLGA) nanoparticle-encapsulated hybrid compound SA-2, possessing nitric oxide (NO) donating and superoxide radical scavenging functionalities. Methods: Previously characterized primary human trabecular meshwork (hTM) cells were used for the study. hTM cells were treated with SA-2 (100 µM, 200 µM, and 1,000 µM), SA-2 PLGA-loaded nanosuspension (SA-2 NPs, 0.1%), or vehicle for 30 min. Cyclic guanosine monophosphate (cGMP) and super oxide dismutase (SOD) levels were analyzed using commercial kits. In another experiment, hTM cells were pretreated with tert-butyl hydrogen peroxide (TBHP, 300 µM) for 30 min followed by treatment with escalating doses of SA-2 for 24 h, and CellTiter 96 cell proliferation assay was performed. For the biodistribution study, the cornea, aqueous humor, vitreous humor, retina, choroid, and sclera were collected after 1 h of administration of a single eye drop (30 µl) of SA-2 NPs (1% w/v) formulated in PBS to rat (n = 6) eyes. Compound SA-2 was quantified using high performance liquid chromatography /mass spectrometry (HPLC/MS). For the IOP-lowering activity study, a single SA-2 NPs (1%) eye drop was instilled in normotensive rats eyes and in the IOP-elevated rat eyes (n = 3/group, in the Morrison model of glaucoma), or Ad5TGFß2-induced ocular hypertensive (OHT) mouse eyes (n = 5/group). IOP was measured at various time points up to 72 h, and the experiment was repeated in triplicate. Mouse aqueous humor outflow facility was determined with multiple flow-rate infusion and episcleral venous pressure estimated with manometry. Results: SA-2 upregulated cGMP levels (six- to ten-fold) with an half maximal effective concentration (EC50) of 20.3 µM in the hTM cells and simultaneously upregulated (40-fold) the SOD enzyme when compared with the vehicle-treated hTM cells. SA-2 also protected hTM cells from TBHP-induced decrease in cell survival with an EC50 of 0.38 µM. A single dose of slow-release SA-2 NPs (1% w/v) delivered as an eye drop significantly lowered IOP (by 30%) in normotensive and OHT rodent eyes after 3 h post-dose, with the effect lasting up to 72 h. A statistically significant increase in aqueous outflow facility and a decrease in episcleral venous pressure was observed in rodents at this dose at 54 h. Conclusions: Hybrid compound SA-2 upregulated cGMP in hTM cells, increased outflow facility and decreased IOP in rodent models of OHT. Compound SA-2 possessing an antioxidant moiety provided additive cytoprotective activity to oxidatively stressed hTM cells by scavenging reactive oxygen species (ROS) and increasing SOD enzyme activity. Additionally, the PLGA nanosuspension formulation (SA-2 NPs) provided longer duration of IOP-lowering activity (up to 3 days) in comparison with the free non-encapsulated SA-2 drug. The data have implications for developing novel, non-prostaglandin therapeutics for IOP-lowering and cytoprotective effects with the possibility of an eye drop dosing regimen of once every 3 days for patients with glaucoma.


Asunto(s)
Antihipertensivos/uso terapéutico , Modelos Animales de Enfermedad , Presión Intraocular/efectos de los fármacos , Hipertensión Ocular/tratamiento farmacológico , Piperidinas/uso terapéutico , Malla Trabecular/efectos de los fármacos , Administración Oftálmica , Adulto , Anciano de 80 o más Años , Animales , Antihipertensivos/farmacocinética , Antihipertensivos/farmacología , Humor Acuoso/fisiología , Disponibilidad Biológica , Células Cultivadas , GMP Cíclico/metabolismo , Portadores de Fármacos , Femenino , Depuradores de Radicales Libres/farmacocinética , Depuradores de Radicales Libres/farmacología , Depuradores de Radicales Libres/uso terapéutico , Glicolatos/química , Humanos , Masculino , Ratones Endogámicos C57BL , Donantes de Óxido Nítrico/farmacocinética , Donantes de Óxido Nítrico/farmacología , Donantes de Óxido Nítrico/uso terapéutico , Hipertensión Ocular/metabolismo , Soluciones Oftálmicas , Piperidinas/farmacocinética , Piperidinas/farmacología , Ratas , Ratas Endogámicas BN , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Esclerótica/irrigación sanguínea , Superóxido Dismutasa/metabolismo , Distribución Tisular , Malla Trabecular/metabolismo , Presión Venosa/fisiología
4.
Sci Rep ; 9(1): 2904, 2019 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-30814582

RESUMEN

In Gram-negative bacterial sepsis, production of excess pro-inflammatory cytokines results in hyperinflammation and tissue injury. Anti-inflammatory cytokines such as IL-10 inhibit inflammation and enhance tissue healing. Here, we report a novel approach to treat septicemia associated with intra-abdominal infection in a murine model by delicately balancing pro- and anti-inflammatory cytokines. A novel oligosaccharide compound AVR-25 selectively binds to the TLR4 protein (IC50 = 0.15 µM) in human peripheral blood monocytes and stimulates IL-10 production. Following the cecal ligation and puncture (CLP) procedure, intravenous dosing of AVR-25 (10 mg/kg, 6-12 h post-CLP) alone and in combination with antibiotic imipenem protected both young adult (10-12 week old) and aged (16-18 month old) mice against polymicrobial infection, organ dysfunction, and death. Proinflammatory cytokines (TNF-α, MIP-1, i-NOS) were decreased significantly and restoration of tissue damage was observed in all organs. A decrease in serum C-reactive protein (CRP) and bacterial colony forming unit (CFU) confirmed improved bacterial clearance. Together, these findings demonstrate the therapeutic ability of AVR-25 to mitigate the storm of inflammation and minimize tissue injury with high potential for adjunctive therapy in intra-abdominal sepsis.


Asunto(s)
Envejecimiento/fisiología , Antiinflamatorios no Esteroideos/uso terapéutico , Quitina/uso terapéutico , Infecciones por Bacterias Gramnegativas/tratamiento farmacológico , Infecciones Intraabdominales/tratamiento farmacológico , Leucocitos Mononucleares/efectos de los fármacos , Oligosacáridos/uso terapéutico , Sepsis/prevención & control , Animales , Ciego/cirugía , Células Cultivadas , Quitina/química , Citocinas/metabolismo , Modelos Animales de Enfermedad , Infecciones por Bacterias Gramnegativas/complicaciones , Humanos , Mediadores de Inflamación/metabolismo , Infecciones Intraabdominales/complicaciones , Leucocitos Mononucleares/fisiología , Ratones , Ratones Endogámicos C57BL , Oligosacáridos/química , Sepsis/etiología , Receptor Toll-Like 4/metabolismo
5.
Nanomedicine ; 6(1): 64-9, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19446653

RESUMEN

A major problem associated with drug therapy is the inability to deliver pharmaceuticals to a specific site of the body without causing nonspecific toxicity. Development of magnetic nanoparticles and techniques for their safe transport and concentration in specific sites in the body would constitute a powerful tool for gene/drug therapy in vivo. Furthermore, drug delivery in vitro could improve further if the drugs were modified with antibodies, proteins, or ligands. For in vivo experiments, magnetic nanoparticles were conjugated with plasmid DNA expressing enhanced green fluorescent protein (EGFP) and then coated with chitosan. These particles were injected into mice through the tail vein and directed to the heart and kidneys by means of external magnets of 25 gauss or 2kA-kA/m. These particles were concentrated in the lungs, heart, and kidneys of mice, and the expression of EGFP in these sites were monitored. The expression of EGFP in specific locations was visualized by whole-body fluorescent imaging, and the concentration of these particles in the designated body locations was confirmed by transmission electron microscopy. In another model system, we used atrial natriuretic peptide and carcinoembryonic antigen antibodies coupled to the chitosan-coated magnetic nanoparticles to target cells in vitro. The present work demonstrates that a simple external magnetic field is all that is necessary to target a drug to a specific site inside the body without the need to functionalize the nanoparticles. However, the option to use magnetic targeting with external magnets on functionalized nanoparticles could prove as a more efficient means of drug delivery. FROM THE CLINICAL EDITOR: This paper addresses targeted drug delivery with magnetic nanoparticles. The authors demonstrate that a simple external magnetic field is sufficient to target a drug to specific sites in the body without the need for functionalized nanoparticles, at least in selected organs and diseases.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Compuestos Férricos/química , Magnetismo/métodos , Nanopartículas/química , Animales , Línea Celular Tumoral , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Ratones , Microscopía Fluorescente , Nanopartículas/ultraestructura , Tamaño de la Partícula , Espectroscopía Infrarroja por Transformada de Fourier , Transfección
6.
Cancer Res ; 68(1): 249-56, 2008 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-18172317

RESUMEN

The receptor for atrial natriuretic peptide (ANP), natriuretic peptide receptor A (NPRA), is expressed in cancer cells, and natriuretic peptides have been implicated in cancers. However, the direct role of NPRA signaling in tumorigenesis remains elusive. Here, we report that NPRA expression and signaling is important for tumor growth. NPRA-deficient mice showed significantly reduced antigen-induced pulmonary inflammation. NPRA deficiency also substantially protected C57BL/6 mice from lung, skin, and ovarian cancers. Furthermore, a nanoparticle-formulated interfering RNA for NPRA attenuated B16 melanoma tumors in mice. Ectopic expression of a plasmid encoding NP73-102, the NH(2)-terminal peptide of the ANP prohormone, which down-regulates NPRA expression, also suppressed lung metastasis of A549 cells in nude mice and tumorigenesis of Line 1 cells in immunocompetent BALB/c mice. The antitumor activity of NP73-102 was in part attributed to apoptosis of tumor cells. Western blot and immunohistochemistry staining indicated that the transcription factor, nuclear factor-kappaB, was inactivated, whereas the level of tumor suppressor retinoblastoma protein was up-regulated in the lungs of NPRA-deficient mice. Furthermore, expression of vascular endothelial growth factor was down-regulated in the lungs of NPRA-deficient mice compared with that in wild-type mice. These results suggest that NPRA is involved in tumor angiogenesis and represents a new target for cancer therapy.


Asunto(s)
Guanilato Ciclasa/fisiología , Neoplasias/irrigación sanguínea , Neovascularización Patológica/genética , Receptores del Factor Natriurético Atrial/fisiología , Adenocarcinoma/genética , Adenocarcinoma/patología , Animales , Apoptosis , Transformación Celular Neoplásica/genética , Femenino , Guanilato Ciclasa/antagonistas & inhibidores , Guanilato Ciclasa/genética , Inflamación/genética , Inflamación/patología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Ratones , Ratones Noqueados , Ratones Mutantes , FN-kappa B/metabolismo , Nanopartículas/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neovascularización Patológica/tratamiento farmacológico , Neoplasias Ováricas/genética , Interferencia de ARN , ARN Interferente Pequeño/genética , Receptores del Factor Natriurético Atrial/antagonistas & inhibidores , Receptores del Factor Natriurético Atrial/genética , Proteína de Retinoblastoma/metabolismo , Neoplasias Cutáneas/genética , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/metabolismo
7.
Allergy Asthma Clin Immunol ; 4(3): 95-105, 2008 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-20525130

RESUMEN

: The use of chitosan nanoparticles as carriers for expression plasmids represents a major improvement in gene expression technology. We demonstrated previously that treatment with chitosan interferon-gamma (IFN-gamma) plasmid deoxyribonucleic acid (DNA) nanoparticles (chitosan interferon-gamma nanogene [CIN]) led to in situ production of IFN-gamma and a reduction in inflammation and airway reactivity in mice, but the mechanism underlying the immunomodulatory effects of CIN remains unclear. In this report, the effect of CIN treatment on the immune responses of CD8+ T cells and dendritic cells was examined in a BALB/c mouse model of ovalbumin (OVA)-induced allergic asthma. OT1 mice (OVA-T cell receptor [TCR] transgenic) were also used to test the effects of CIN on OVA-specific CD8+ T cells. CIN treatment caused a reduction in IFN-gamma production in a subpopulation of OVA-specific CD8+ T cells cultured in vitro in the presence of OVA. CIN also reduced apoptosis of the CD8+ T cells. Examination of dendritic cells from lung and lymph nodes indicated that CIN treatment decreased their antigen-presenting activity, as evident from the reduction in CD80 and CD86 expression. Furthermore, CIN treatment significantly decreased the number of CD11c+b+ dendritic cells in lymph nodes, suggesting that endogenous IFN-gamma expression may immunomodulate dendritic cell migration and activation. CIN therapy results in a reduction in proinflammatory CD8+ T cells and decreases the number and antigen-presenting activity of dendritic cells.

8.
Nanomedicine ; 3(2): 132-7, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17572355

RESUMEN

Novel hybrid nanoparticles comprised of hyaluronic acid (HA) and iron oxide were synthesized and characterized for the first time with the average diameter of less than 160 nm. The iron oxide (Fe2O3) particles are hybridized between HA layers by electrostatic interactions between the positive surface charge of the Fe2O3 nanoparticles and the negative charge of the carboxylate groups of HA, forming a corral-like structure. The particles were also characterized by FTIR and NMR to verify the hybridization. The particles were tested for their ability to deliver peptides to the cells using HEK293 and A549 cells. Results show that these particles delivered peptides at about 100% level. These HA-iron oxide nanoparticles are expected to be useful in developing effective tissue and cell targeting systems.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Compuestos Férricos/química , Ácido Hialurónico/química , Riñón/metabolismo , Neoplasias Pulmonares/metabolismo , Magnetismo/uso terapéutico , Nanopartículas/administración & dosificación , Péptidos/administración & dosificación , Péptidos/farmacocinética , Línea Celular , Humanos , Ensayo de Materiales , Nanopartículas/química
9.
Virol J ; 3: 32, 2006 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-16719922

RESUMEN

BACKGROUND: Respiratory syncytial virus (RSV) infection is the major cause of bronchiolitis in infants and is a risk factor for the development of asthma. Allergic asthmatics are more susceptible to RSV infection and viral exacerbation. METHODS: Since the effectiveness of corticosteroids in treating RSV infection has been controversial, we tested fluticasone propionate (FP) and salmeterol (Sal) alone versus FP plus Sal (FPS) on RSV-induced airway inflammation. Mice were sensitized and challenged with ovalbumin (OVA) and infected with RSV. Following infection they were treated with FP, Sal, or FPS intranasally and airway hyperreactivity (AHR), inflammation and RSV titers were examined. RESULTS: The group treated with FPS showed significantly lower AHR compared to the group treated with FP or Sal alone. The group treated with FP alone showed slightly decreased (non-significant) AHR compared to controls. Treatment with FPS resulted in significant decreases in the percentage of eosinophils and neutrophils in bronchoalveolar lavage fluid and in lung pathology compared to FP or Sal. FP alone decreased eosinophils but not neutrophils or lymphocytes, while Sal alone decreased eosinophils and neutrophils but not lymphocytes. FPS treatment of mice infected with RSV in the absence of allergen sensitization resulted in a 50% decrease of RSV titer in the lung and a reduction in neutrophils compared to FP or Sal. CONCLUSION: Together, these results indicate that fluticasone in combination with salmeterol is a more effective treatment for decreasing airway hyperreactivity and inflammation than either of them alone in allergen-sensitized, RSV-infected mice.


Asunto(s)
Albuterol/análogos & derivados , Alérgenos/inmunología , Androstadienos/administración & dosificación , Androstadienos/uso terapéutico , Ovalbúmina/inmunología , Infecciones por Virus Sincitial Respiratorio/tratamiento farmacológico , Albuterol/administración & dosificación , Albuterol/uso terapéutico , Animales , Antialérgicos/administración & dosificación , Antialérgicos/uso terapéutico , Asma/tratamiento farmacológico , Lavado Broncoalveolar , Broncodilatadores/administración & dosificación , Broncodilatadores/uso terapéutico , Combinación de Medicamentos , Femenino , Fluticasona , Combinación Fluticasona-Salmeterol , Pulmón/efectos de los fármacos , Pulmón/patología , Ratones , Ratones Endogámicos BALB C , Infecciones por Virus Sincitial Respiratorio/inmunología , Xinafoato de Salmeterol
10.
Nat Med ; 11(1): 56-62, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15619625

RESUMEN

Respiratory syncytial virus (RSV) infection is one of the major causes of respiratory tract infection for which no vaccine or antiviral treatment is available. The RSV NS1 protein seems to antagonize the host interferon (IFN) response; however, its mechanism is unknown. Here, we used a plasmid-borne small interfering RNA targeting the NS1 gene (siNS1) to examine the role of NS1 in modulating RSV infection. RSV replication was reduced in A549 cells, but not IFN-deficient Vero cells, transfected with siNS1. siNS1 induced upregulated expression of IFN-beta and IFN-inducible genes in A549 cells. siNS1-transfected human dendritic cells, upon RSV infection, produced elevated type-1 IFN and induced differentiation of naive CD4+ T cells to T helper type 1 (TH1) cells. Mice treated intranasally with siNS1 nanoparticles before or after infection with RSV showed substantially decreased virus titers in the lung and decreased inflammation and airway reactivity compared to controls. Thus, siNS1 nanoparticles may provide an effective inhibition of RSV infection in humans.


Asunto(s)
Antivirales/farmacología , Células Dendríticas/metabolismo , ARN Interferente Pequeño/farmacología , Virus Sincitiales Respiratorios/efectos de los fármacos , Proteínas no Estructurales Virales/genética , Humanos , Interferón Tipo I/metabolismo , Pulmón/patología , Pulmón/virología , Nanoestructuras , Infecciones por Virus Sincitial Respiratorio/prevención & control , Virus Sincitiales Respiratorios/genética , Células TH1/metabolismo , Regulación hacia Arriba , Proteínas no Estructurales Virales/metabolismo
11.
J Allergy Clin Immunol ; 113(1): 79-85, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14713911

RESUMEN

BACKGROUND: The natriuretic hormone peptide (NHP)(99-126), a C-terminal peptide of pro-atrial natriuretic factor (proANF), induces bronchodilatory effects in people with asthma. Recently, another plasmid-encoded C-terminal peptide, pNHP(73-102), was shown to induce a long-lasting bronchoprotective effect in a mouse model of allergic asthma. OBJECTIVE: This study was carried out to determine the role of lung epithelial cells in the bronchoprotective and anti-inflammatory activity of these peptides. METHODS: Human type II alveolar epithelial cells (A549) and normal human bronchial epithelial (NHBE) cells were transfected with pNHP(73-102) to test the effect of this peptide on activation of these cells. After transfection, cells were analyzed for changes in Ca(++) and nitric oxide (NO) levels. Also, activation of NFkappaB and the extracellularly regulated kinase (ERK) 1, 2 signaling pathway was examined by luciferase reporter assay and phosphorylation studies respectively. RESULTS: Analysis of intracellular Ca(++) levels in pNHP(73-102) -transfected A549 or NHBE showed that the peptide increases release. This Ca(++) release was accompanied by an increase in the production of NO. Also, overexpression of pNHP(73-102), but not pVAX control, in phorbol myristate acetate-activated A549 cells resulted in a significant decrease in expression of a cotransfected nuclear factorkappaB (NFkappaB)-luciferase reporter. Similarly, pNHP(73-102) decreased TNF-alpha-induced NFkappaB activation in NHBE cells. Furthermore, NHP(73-102) but not atrial natriuretic peptide decreased phosphorylation of Erk-1, 2 in A549 cells. CONCLUSIONS: Overexpression of pNHP(73-102) in epithelial cells causes increased production of intracellular Ca(++) and NO, with a concomitant decrease in activation of NFkappaB and ERK1, 2. These results suggest a bronchodilatory and anti-inflammatory activity of this peptide.


Asunto(s)
Factor Natriurético Atrial/fisiología , Células Epiteliales/metabolismo , Alveolos Pulmonares/citología , Factor Natriurético Atrial/genética , Calcio/metabolismo , Línea Celular , Humanos , Immunoblotting , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico Sintasa de Tipo III , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...