Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 5: 16331, 2015 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-26573509

RESUMEN

MicroRNAs were described to target mRNA and regulate the transcription of genes involved in processes de-regulated in tumorigenesis, such as proliferation, differentiation and survival. In particular, the miRNA let-7 has been suggested to regulate the expression of the KRAS gene, a common mutated gene in non-small cell lung cancer (NSCLC), through a let-7 complementary site (LCS) in 3'UTR of KRAS mRNA. We have reported the analysis performed on the role of the polymorphism located in the KRAS-LCS (rs61764370) which is involved in the disruption of the let-7 complementary site in NSCLC patients enrolled within the TAILOR trial, a randomised trial comparing erlotinib versus docetaxel in second line treatment. In our cohort of patients, KRAS-LCS6 polymorphism did not have any impact on both overall survival (OS) and progression free survival (PFS) and was not associated with any patient's baseline characteristics included in the study. Overall, patients had a better prognosis when treated with docetaxel instead of erlotinib for both OS and PFS. Considering KRAS-LCS6 status, the TG/GG patients had a benefit from docetaxel treatment (HR(docetaxel vs erlotinib) = 0.35, 95% CI 0.15-0.79, p = 0.011) compared with the TT patients (HR(docetaxel vs erlotinib) = 0.72, 95% CI 0.52-1.01, p = 0.056) in terms of PFS.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Clorhidrato de Erlotinib/uso terapéutico , Neoplasias Pulmonares/tratamiento farmacológico , Taxoides/uso terapéutico , Proteínas ras/genética , Regiones no Traducidas 3' , Anciano , Alelos , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/mortalidad , Supervivencia sin Enfermedad , Docetaxel , Femenino , Genotipo , Humanos , Estimación de Kaplan-Meier , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/mortalidad , Masculino , MicroARNs/genética , MicroARNs/metabolismo , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple , Modelos de Riesgos Proporcionales
2.
J Gene Med ; 14(12): 733-45, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23074129

RESUMEN

BACKGROUND: Epithelial sodium channel (ENaC) hyperactivity has been implicated in the pathogenesis of cystic fibrosis (CF) by dysregulation of fluid and electrolytes in the airways. In the present study, we show proof-of-principle for ENaC inhibition by lentiviral-mediated RNA interference. METHODS: Immortalized normal (H441) and CF mutant (CFBE) airway cells, and differentiated human bronchial epithelial cells in air liquid interface culture (HBEC-ALI) were transduced with a vesicular stomatitis virus G glycoprotein pseudotyped lentiviral (LV) vector expressing a short hairpin RNA (shRNA) targeting the α subunit of ENaC (ENaCα), and a marker gene. Efficacy of ENaCα down-regulation was assayed by the real-time polymerase chain reaction (PCR), membrane potential assay, western blotting, short-circuit currents and fluid absorption. Off-target effects were investigated by a lab-on-a-chip quantitative PCR array. RESULTS: Transduction to near one hundred percentage efficiency of H441, CFBE and HBEC-ALI was achieved by the addition of the LV vector before differentiation and polarization. Transduction resulted in the inhibition of ENaCα mRNA and antigen expression, and a proportional decrease in ENaC-dependent short circuit current and fluid transport. No effect on transepithelial resistance or cAMP-induced secretion responses was observed in HBEC-ALI. The production of interferon α and pro-inflammatory cytokine mRNA, indicating Toll-like receptor 3 or RNA-induced silencing complex mediated off-target effects, was not observed in HBEC-ALI transduced with this vector. CONCLUSIONS: We have established a generic method for studying the effect of RNA interference in HBEC-ALI using standard lentiviral vectors. Down-regulation of ENaCα by lentiviral shRNA expression vectors as shown in the absence off-target effects has potential therapeutic value in the treatment of cystic fibrosis.


Asunto(s)
Células Epiteliales/metabolismo , Canales Epiteliales de Sodio/genética , Vectores Genéticos , Lentivirus/genética , ARN Interferente Pequeño/genética , Mucosa Respiratoria/metabolismo , Línea Celular , Células Epiteliales/virología , Canales Epiteliales de Sodio/metabolismo , Regulación de la Expresión Génica , Orden Génico , Técnicas de Transferencia de Gen , Humanos , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Mucosa Respiratoria/virología , Transducción Genética
3.
Med Sci Monit ; 16(12): BR361-6, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21119568

RESUMEN

BACKGROUND: Pseudomonas aeruginosa is an opportunistic pathogen that causes chronic endobronchial infections in cystic fibrosis (CF) patients. The role of bacterial internalization in the clearance of P. aeruginosa from the airways is controversial. MATERIAL/METHODS: A xenograft model was used to study P. aeruginosa strain PAO-1 clearance and internalization by the human airways in vivo. Human lung and tracheal rudiments, obtained from therapeutic abortions (20±2 weeks of gestation), were subcutaneously implanted in the flanks of SCID mice and infected after 14-22 weeks of engraftment. Lungs were surgically exposed and P. aeruginosa was injected in the pulmonary parenchima. Opercula closing the tracheal openings were excised, mucus removed, and bacterial inoculum was injected into the lumen. Internalization was studied at 4 hours post-infection on single-cell suspensions, while clearance was evaluated after 24-72 hours from the infection on homogenized tissues. RESULTS: Tracheae and lungs were morphologically identical to the adult human tissues, as evaluated by standard histology. Both types of xenografts showed a very low level of bacterial internalization (0.004-0.25% of total recovered bacteria), although tracheal xenografts presented more than 100 times greater internalization than did lung xenografts. Both lung and tracheal xenografts did not clear the injected bacteria for each inocolum, even at very low doses (100 colony forming units). CONCLUSIONS: P. aeruginosa internalization by epithelial cells occurs, albeit at very low levels, and is not sufficient to clear bacteria in the airway xenograft model. This model could be used for studying chronic respiratory infections in CF patients.


Asunto(s)
Células Epiteliales/microbiología , Modelos Animales , Pseudomonas aeruginosa/fisiología , Infecciones del Sistema Respiratorio/microbiología , Trasplante Heterólogo , Animales , Trasplante de Tejido Fetal , Humanos , Trasplante de Pulmón , Ratones , Ratones SCID , Tráquea/microbiología , Tráquea/trasplante
4.
J Gene Med ; 12(9): 747-54, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20821745

RESUMEN

BACKGROUND: Lentiviral (LV) vectors are able to only slowly and inefficiently transduce nondividing cells such as those of the airway epithelium. To address this issue, we have exploited the magnetofection technique in in vitro models of airway epithelium. METHODS: Magnetofectins were formed by noncovalent interaction between LV particles and polycation-coated iron oxide nanoparticles. Efficiency of LV-mediated transduction (as evaluated through green fluorescent protein (GFP) expression by cytofluorimetric analysis) was measured in bronchial epithelial cells in the presence or absence of a magnetic field. Cytotoxicity was evaluated by lactate dehydrogenase (LDH) release; cell monolayer integrity by measurement of transepithelial resistance (TER) and evaluation of correct zonula occludens-1 (ZO-1) localization at tight junctions (TJs) by immunofluorescence and confocal microscopy. RESULTS: In nonpolarized cells, magnetofectins enhanced LV-mediated transduction at multiplicity of infection (MOI) of 50 up to 3.9-fold upon a 24-h incubation, to levels that approached those achieved at MOI of 200 for LV alone, in the presence or absence of the magnetic field. Magnetofection significantly increased the percentage of transduced cells up to 186-fold already after 15 min of incubation. In polarized cells, magnetofection increased GFP+ cells up to 24-fold compared to LV alone. Magnetofection did not enhance LDH release and slightly altered TER but not ZO-1 localization at the TJs. CONCLUSIONS: We conclude that magnetofection can facilitate in vitro LV-mediated transduction of airway epithelial cells, in the absence of overt cytotoxicity and maintaining epithelial integrity, by lowering the necessary vector dose and reducing the incubation time required to achieve efficient transduction.


Asunto(s)
Células Epiteliales/metabolismo , Técnicas de Transferencia de Gen , Lentivirus/genética , Magnetismo , Mucosa Respiratoria/metabolismo , Polaridad Celular , Supervivencia Celular , Células Cultivadas , Vectores Genéticos/administración & dosificación , Proteínas Fluorescentes Verdes/metabolismo , Humanos , L-Lactato Deshidrogenasa/metabolismo , Proteínas de la Membrana/metabolismo , Fosfoproteínas/metabolismo , Proteína de la Zonula Occludens-1
5.
Viruses ; 2(8): 1577-1588, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21994695

RESUMEN

The aim of this work was to evaluate the efficiency and duration of gene expression mediated by a VSV-G pseudotyped last generation lentiviral (LV) vector. We studied LV efficiency in ex-vivo models of respiratory epithelial cells, obtained from bronchial biopsies and nasal polyps, by GFP epifluorescence and cytofluorimetry. In vivo efficiency and persistence of gene expression was investigated by GFP immunohistochemistry and luciferase activity in lung cryosections and homogenates, respectively, upon intranasal and intratracheal administration protocols in C57Bl/6 mice. Both primary bronchial and nasal epithelial cells were transduced up to 70-80% 72 hr after the LV infection. In vivo nasal luciferase expression was increased by lysophosphatidylcholine pre-treatment of the nose. Conversely, the bronchial epithelium was transduced in the absence of any pre-conditioning treatment and luciferase expression lasted for at least 6 months without any decline. We conclude that a last generation LV vector is a promising gene transfer agent in the target organ of genetic and acquired lung diseases, as in the case of cystic fibrosis.

6.
Virus Res ; 144(1-2): 8-17, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19447514

RESUMEN

Lentiviruses (LVs) are considered one of the most promising tools for gene transfer, however, their potential to induce pro-inflammatory cytokines on delivery into the respiratory tissue remains to be established. Here we tested a third-generation vesicular stomatitis virus (VSV)-G pseudotyped LV vector in the two respiratory epithelial cell lines A549 and CFT1-C2. We observed that the VSV-G LV vector does not induce (a) activation of the nuclear factor (NF)-kappaB, which intervenes in transcription of pro-inflammatory genes; (b) expression of ICAM-1; and (c) transcription of a panel of cytokines, with the exception of a mild and transient (24h) increase of IFN-gamma mRNA. In contrast, an adenovirus-derived vector strongly activated NF-kappaB and different transcripts such as those of ICAM-1, IL-8, RANTES, IP-10, TNF-alpha, IL-6, IL-1 beta. In conclusion, this third-generation VSV-G pseudotyped LV vector does not elicit major pro-inflammatory signals in human airway epithelial cells and appears to be better suited for gene delivery strategies.


Asunto(s)
Células Epiteliales/inmunología , Células Epiteliales/virología , Vectores Genéticos/inmunología , Lentivirus/inmunología , Vesiculovirus/inmunología , Adenoviridae/genética , Adenoviridae/inmunología , Línea Celular , Citocinas/biosíntesis , Perfilación de la Expresión Génica , Humanos , Molécula 1 de Adhesión Intercelular/biosíntesis , Lentivirus/genética , FN-kappa B/biosíntesis , Vesiculovirus/genética
7.
J Gene Med ; 10(12): 1294-302, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18816484

RESUMEN

BACKGROUND: The involvement of surface molecules in HIV-1-derived lentivirus (LV)-mediated transduction of airway epithelial cells has not been studied so far. The present study aimed to evaluate the role of glycosaminoglycans (GAGs) in gene transfer mediated by a third generation vesicular stomatitis virus G glycoprotein (VSV-G) pseudotyped LV vector in an in vitro model of polarized airway epithelial cells. METHODS: Human bronchial (16HBE-S1) and tracheal (CFT1-C2) epithelial cells were grown either on plastic or on filters and transduced with the LV vector polypurine tract (PPT)-green fluoresecent protein (GFP). Zonula Occludens (ZO)-1, a marker of tight junction, and GAG localization were assessed by cytofluorimetry and confocal microscopy. Soluble GAGs and removal of cell surface GAGs were used to affect LV-mediated transduction. RESULTS: Extensive optimization of experimental parameters (presence of polybrene during the infection, the incubation time in the presence of LV particles, period of time intercurring between infection and gene expression analysis) was carried out in plastic-adherent cells. Polybrene resulted to be cytotoxic and was not further used. In CFT1-C2 polarized cells, EGTA treatment determined a 20% decrease in transepithelial resistance, a diminished ZO-1 localization at the tight junction location and a 31% increase in GFP positive cells. Heparane sulfate was distributed evenly on the cell surface. Heparin and soluble chondroitin sulfate A and B inhibited LV-mediated transduction in a dose-dependent fashion. These results were confirmed upon enzymatic removal of GAGs from the cell surface. CONCLUSIONS: Taken together, these results show that GAGs are involved in VSV-G LV transduction of airway epithelial cells.


Asunto(s)
Células Epiteliales/metabolismo , Glicosaminoglicanos/metabolismo , Lentivirus/genética , Glicoproteínas de Membrana/genética , Mucosa Respiratoria/metabolismo , Proteínas del Envoltorio Viral/genética , Animales , Células Cultivadas , Células Epiteliales/citología , Técnicas de Transferencia de Gen , Vectores Genéticos/genética , Glicosaminoglicanos/análisis , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Lentivirus/metabolismo , Ratones , Células 3T3 NIH , Mucosa Respiratoria/citología , Uniones Estrechas/metabolismo , Transducción Genética
8.
Biomacromolecules ; 9(3): 859-66, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18275149

RESUMEN

Efficient gene transfer to the airways by nonviral vectors is a function of different parameters, among which the size and the charge of the transfecting particles. The aim of this study was to determine the transfection efficiency of polyethylenimine (PEI)/albumin polyplexes in ex vivo and in vivo models of respiratory epithelium and to correlate it with biophysical characteristics of the particles. Complexes were obtained by adding different amounts of human serum albumin (HSA) to PEI polyplexes preformed in saline. The presence of HSA caused the formation of bigger and more negative polyplexes and increased PEI transfection efficiency in primary respiratory epithelial cells by 4-6-fold. For in vivo administration to the lung, PEI polyplexes were formed in water and optimized with respect to the N/ P ratio. PEI/pC-Luc complexes gave the highest luciferase expression at N/ P 15 when administered through the trachea. At this N/ P ratio, the size and the surface charge of albumin-containing polyplexes were not different as compared with plain PEI polyplexes. Formulation of PEI polyplexes in the presence of HSA or murine serum albumin (MSA) resulted in a 2-fold increase in luciferase expression. In mice treated with PEI or PEI/MSA polyplexes containing the nuclear beta-gal gene, X-gal staining revealed that transfected cells localized at the bronchiolar epithelium and that PEI/MSA transfected four times as many cells as PEI ( p < 0.05). Finally, double administration of PEI/MSA polyplexes resulted in a further enhancement of transfection of the lung. Our data show that serum albumin enhances PEI-mediated gene transfer to airway epithelial cells in vivo, likely facilitating the uptake of polyplexes, and indicate that this formulation would fulfill the requirement of repeated administration, as necessary in chronic lung diseases like cystic fibrosis.


Asunto(s)
Técnicas de Transferencia de Gen , Vectores Genéticos/metabolismo , Polietileneimina/metabolismo , Mucosa Respiratoria/efectos de los fármacos , Albúmina Sérica/farmacología , Animales , Fenómenos Biofísicos , Biofisica , Electricidad , Humanos , Luciferasas/genética , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Ratones , Polietileneimina/química , Mucosa Respiratoria/metabolismo , Albúmina Sérica/química , Tráquea/efectos de los fármacos , Tráquea/metabolismo , Agua/química , Agua/metabolismo
9.
J Cyst Fibros ; 5(2): 113-9, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16481223

RESUMEN

BACKGROUND: It is not clear whether cystic fibrosis (CF) airway inflammation is a consequence of bacterial infection or is intrinsically dysregulated. The aim of this study was to investigate IL-8 secretion and NF-kappaB activity in primary respiratory epithelial cells cultured from nasal polyps obtained from CF and non-CF subjects. METHODS: NF-kappaB activity was studied by electrophoretic mobility-shift and quantitative colorimetric assays in nuclear extracts. Immunoreactive IL-8 levels were assessed by ELISA in cell culture supernatants. Both parameters were studied at baseline and following challenge with Pseudomonas aeruginosa or stimulation with pro-inflammatory cytokines. RESULTS: Under basal conditions, CF cells presented a significant higher activity of NF-kappaB than non-CF cells (P=0.0004). P. aeruginosa challenge and IL-1beta/H2O2 co-stimulation caused four and two fold induction of NF-kappaB activity in non-CF and CF cells, respectively. IL-8 levels in unstimulated CF cells were significantly higher than in non-CF cells (P=0.0025). Upon incubation with P. aeruginosa and IL-1beta/H2O2, non-CF cells produced 6.3 times more IL-8 than unstimulated cells, whereas IL-8 secretion increased only of 1.4 times in CF cells. CONCLUSIONS: CF respiratory epithelial cells exhibit a basal dysregulated production of IL-8 that partially correlates to enhanced NF-kappaB activity. Our data corroborate the hypothesis of a basal exaggerated inflammatory response in the CF respiratory epithelium.


Asunto(s)
Fibrosis Quística/metabolismo , Interleucina-8/metabolismo , FN-kappa B/metabolismo , Mucosa Nasal/metabolismo , Células Cultivadas , Humanos , Peróxido de Hidrógeno/farmacología , Interleucina-1/farmacología , Mucosa Nasal/efectos de los fármacos , Mucosa Nasal/microbiología , Pseudomonas aeruginosa/fisiología
10.
J Gene Med ; 7(12): 1555-64, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16028303

RESUMEN

BACKGROUND: The interaction of polyethylenimine (PEI) polyplexes with proteins in cystic fibrosis (CF) airway secretions poses a significant hurdle to this nonviral delivery system. The aim of this study was to evaluate whether albumin may increase the efficiency of PEI complexes in mediating gene transfer into respiratory epithelial cells in the presence of CF mucus. METHODS: PEI (25 kDa) was complexed to DNA in the presence of human serum albumin (HSA) and used to transfect confluent A549 and 9HTEo- cells. Alternatively, albumin was added to preformed PEI-DNA complexes. The cytotoxicity of complexes was analysed by the LDH (lactate dehydrogenase) assay. CF CFT1-C2 cells were allowed to polarise and were transfected either with luciferase- or CFTR-expressing plasmids. To evaluate the effect of CF respiratory secretions on transfection efficiency, confluent cells were transfected in the presence of sputum obtained from two CF patients. RESULTS: The ternary PEI-HSA complexes increased luciferase expression in confluent cultures in a dose-dependent fashion up to 100 times as compared to PEI-DNA. The number of GFP-expressing cells, as evaluated by epifluorescence, was augmented several-fold. When HSA was added to preformed PEI-DNA complexes, a further 5-10-fold increase in gene expression was observed. No significant cytotoxicity was observed with either PEI or PEI-HSA polyplexes. The ternary complexes determined detectable CFTR gene transfer and expression at the apical membrane in polarised CFT1-C2 cells, as evaluated by confocal microscopy. CF sputum inhibited PEI-mediated gene transfer by 7-186-fold. Although luciferase expression mediated by PEI-HSA was still inhibited by CF sputum, these levels were 18-83.8-fold higher than with PEI. CONCLUSIONS: Our results demonstrate that albumin increases PEI gene transfer efficiency in confluent and polarised respiratory epithelial cells and can allow CFTR gene expression in the appropriate cellular compartment. PEI-HSA complexes display a higher efficiency than PEI also in the presence of CF sputum, indicating that albumin-containing polyplexes may help overcome barriers imposed by CF airway secretions.


Asunto(s)
Fibrosis Quística/terapia , ADN/metabolismo , Terapia Genética/métodos , Polietileneimina/metabolismo , Mucosa Respiratoria/metabolismo , Albúmina Sérica/metabolismo , Transfección/métodos , Línea Celular , Expresión Génica/efectos de los fármacos , Proteínas Fluorescentes Verdes/metabolismo , Humanos , L-Lactato Deshidrogenasa , Luciferasas/metabolismo , Microscopía Fluorescente , Albúmina Sérica/farmacología , Esputo/metabolismo , Transgenes/genética
11.
Pediatr Pulmonol ; 40(1): 15-21, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15858808

RESUMEN

It is not known whether cytokine levels in sputum may be used as outcome measures after parenteral antibiotic therapy in cystic fibrosis (CF) patients. Here, we assessed the effects of antibiotic therapy on cytokine levels in sputum and serum obtained from young CF patients. Thirty-two CF patients (14 females; mean age, 18.6 years; range, 11.4-35.7 years), consecutively admitted at the CF Center of Milan for parenteral antibiotic therapy during pulmonary exacerbation, were enrolled in the study. Before and after 21 days (range, 5-41) of intravenous antibiotic treatment, all patients underwent routine laboratory determinations (including white blood cell (WBC) count and C-reactive protein (CRP)), a chest X-ray, pulmonary function tests (forced expiratory volume in 1 sec (FEV1) and forced vital capacity (FVC) as % predicted), and sputum cultures. Interleukin (IL)-6, IL-8, IL-10, and tumor necrosis factor (TNF)-alpha levels in serum and sputum samples were determined by means of immunometric assays. After therapy, FEV1 and FVC significantly improved (median increase of 7.5% and 8.5% predicted, respectively), while CRP and WBC count were significantly decreased (median values from 14 to 5.5 mg/dl and from 8,350 to 7,400 n/mm3, respectively). While levels of IL-6 and IL-10 in sputum were generally undetectable, IL-8 and TNF-alpha were always measurable, and IL-8 levels significantly decreased after antibiotic treatment (median values from 7,165 to 5,415 pg/ml). Following antibiotic therapy, IL-8 and TNF-alpha levels in sputum were inversely related with both FEV(1) and FVC. In conclusion, TNF-alpha and IL-8 levels in sputum of young CF patients with pulmonary exacerbation were always detectable and may be useful, noninvasive outcome measures to assess response to therapy in CF patients.


Asunto(s)
Antibacterianos/uso terapéutico , Fibrosis Quística/complicaciones , Fibrosis Quística/metabolismo , Citocinas/metabolismo , Infecciones del Sistema Respiratorio/tratamiento farmacológico , Infecciones del Sistema Respiratorio/metabolismo , Esputo/metabolismo , Adolescente , Adulto , Biomarcadores/metabolismo , Niño , Fibrosis Quística/inmunología , Femenino , Humanos , Inflamación/etiología , Inflamación/metabolismo , Masculino , Pruebas de Función Respiratoria , Infecciones del Sistema Respiratorio/diagnóstico , Infecciones del Sistema Respiratorio/etiología
12.
J Cyst Fibros ; 3 Suppl 2: 197-201, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15463958

RESUMEN

Cystic fibrosis (CF) airway becomes colonized with only a limited number of bacterial pathogens. It is of paramount importance to establish in vitro and in vivo models to better understand bacterial-host interactions under CF-like conditions. In this article, in vitro methods suitable to study Pseudomonas aeruginosa (Pa) and Staphylococcus aureus (Sa) adherence to and uptake by airway epithelial cells are described. Acute and chronic respiratory infection models, which have been used in CF transgenic mice and mimic human CF lung pathology, are also taken into consideration.


Asunto(s)
Infecciones Bacterianas/fisiopatología , Pseudomonas aeruginosa/fisiología , Mucosa Respiratoria/fisiología , Infecciones del Sistema Respiratorio/fisiopatología , Staphylococcus aureus/fisiología , Animales , Apoptosis/fisiología , Adhesión Bacteriana/fisiología , Técnicas de Cultivo de Célula , Fibrosis Quística/fisiopatología , Fragmentación del ADN/fisiología , Endocitosis/fisiología , Células Epiteliales/fisiología , Humanos , Ratones , Ratones Transgénicos , Modelos Biológicos , Mucosa Respiratoria/fisiopatología
13.
J Cyst Fibros ; 2(3): 129-35, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15463861

RESUMEN

The pathological hallmark of cystic fibrosis (CF) chronic inflammatory response is the massive neutrophil influx into the airways. This dysregulated neutrophil emigration may be caused by the abnormal secretion of chemoattractants by respiratory epithelial cells and polarised lymphocyte T-helper response. Neutrophils from CF patients have a different response to inflammatory mediators than neutrophils from normal subjects, indicating that they are primed in vivo before entering the CF airways. CF neutrophils secrete more myeloperoxidase and elastase, mobilise less opsonin receptors and release less L-selectin than non-CF neutrophils. Moreover, they show altered cytokine production and a dysregulated chemotaxis response. Laboratory studies now suggest that CFTR is involved in regulating some neutrophil functions and indicate that altered properties of CF neutrophils may depend on genetic factors. Current gene therapy approaches are targeted to the respiratory epithelium, but many hurdles oppose an efficient and efficacious CFTR gene transfer. The possibility of CFTR gene therapy-based approach targeting CF neutrophils at the hematopoietic stem cell level is discussed.


Asunto(s)
Fibrosis Quística/fisiopatología , Neutrófilos/fisiología , Bronquios/fisiopatología , Movimiento Celular/fisiología , Enfermedad Crónica , Fibrosis Quística/terapia , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Epitelio/fisiopatología , Terapia Genética , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...