Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Curr Gene Ther ; 15(6): 581-90, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26471068

RESUMEN

A key challenge in pulmonary gene therapy for cystic fibrosis is to provide long-term correction of the genetic defect. This may be achievable by targeting airway epithelial stem/progenitor cells with an integrating vector. Here, we evaluated the ability of a lentiviral vector, derived from the simian immunodeficiency virus and pseudotyped with F and HN envelope proteins from Sendai virus, to transduce progenitor basal cells of the mouse nasal airways. We first transduced basal cell-enriched cultures ex vivo and confirmed efficient transduction of cytokeratin-5 positive cells. We next asked whether progenitor cells could be transduced in vivo. We evaluated the transduction efficiency in mice pretreated by intranasal administration of polidocanol to expose the progenitor cell layer. Compared to control mice, polidocanol treated mice demonstrated a significant increase in the number of transduced basal cells at 3 and 14 days post vector administration. At 14 days, the epithelium of treated mice contained clusters (4 to 8 adjacent cells) of well differentiated ciliated, as well as basal cells suggesting a clonal expansion. These results indicate that our lentiviral vector can transduce progenitor basal cells in vivo, although transduction required denudation of the surface epithelium prior to vector administration.


Asunto(s)
Fibrosis Quística/terapia , Células Epiteliales/citología , Terapia Genética/métodos , Mucosa Respiratoria/citología , Células Madre/citología , Tráquea/citología , Animales , Células Cultivadas , Fibrosis Quística/genética , Femenino , Vectores Genéticos/genética , Proteínas Fluorescentes Verdes/genética , Lentivirus/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Polidocanol , Polietilenglicoles/farmacología , Virus Sendai/genética , Virus de la Inmunodeficiencia de los Simios/genética , Transducción Genética
2.
Cochrane Database Syst Rev ; (6): CD011079, 2015 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-26041053

RESUMEN

BACKGROUND: Humidified high flow nasal cannula (HHFNC) delivers humidified gas at increased flow rates via binasal prongs and is becoming widely accepted as a method of non-invasive respiratory support for preterm infants. While indications for the use of (HHFNC) and its associated risks and benefits are being investigated, the best strategy for the discontinuation of HHFNC remains unknown. At what point an infant is considered stable enough to attempt to start withdrawing their HHFNC is not known. The criteria for a failed attempt at HHFNC discontinuation is also unclear. OBJECTIVES: To determine the risks and benefits of different strategies used for the discontinuation of HHFNC in preterm infants. SEARCH METHODS: We searched the Cochrane Neonatal Review Group Specialized Register, PubMed (1966 to March 2015), CINAHL (1982 to March 2015), EMBASE (1980 to March 2015), and the Cochrane Central Register of Controlled Trials (CENTRAL). Also, we checked previous reviews, including cross references. We searched for following web sites for ongoing trials: ClinicalTrials.gov and controlled-trials.com. SELECTION CRITERIA: We included randomised controlled trials (RCTs) and quasi-RCTs in which either individual newborn infants or clusters of infants (such as separate neonatal units) were randomised to different HHFNC withdrawal strategies (from the first time they come off HHFNC and any subsequent weaning, or withdrawal attempt, or both). DATA COLLECTION AND ANALYSIS: We used standard methods of Cochrane and the Cochrane Neonatal Review Group. MAIN RESULTS: We identified no eligible studies examining the best strategy to wean or withdraw HHFNC once started as respiratory support in preterm infants AUTHORS' CONCLUSIONS: There is currently no evidence available to suggest the best strategy for weaning and withdrawing HHFNC as a respiratory support in preterm infants. Research is required into the best strategy for withdrawal of HHFNC and to which subgroups this applies. Clear criteria for the definition of stability prior to attempting to withdraw HHFNC needs to be established. Furthermore, clear definitions are needed as to what constitutes failure of HHFNC.


Asunto(s)
Presión de las Vías Aéreas Positiva Contínua/métodos , Remoción de Dispositivos/métodos , Recien Nacido Prematuro , Ventilación no Invasiva/métodos , Desconexión del Ventilador/métodos , Remoción de Dispositivos/efectos adversos , Humanos , Humidificadores , Recién Nacido , Desconexión del Ventilador/efectos adversos
3.
Am J Respir Crit Care Med ; 186(9): 846-56, 2012 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-22955314

RESUMEN

RATIONALE: Ongoing efforts to improve pulmonary gene transfer thereby enabling gene therapy for the treatment of lung diseases, such as cystic fibrosis (CF), has led to the assessment of a lentiviral vector (simian immunodeficiency virus [SIV]) pseudotyped with the Sendai virus envelope proteins F and HN. OBJECTIVES: To place this vector onto a translational pathway to the clinic by addressing some key milestones that have to be achieved. METHODS: F/HN-SIV transduction efficiency, duration of expression, and toxicity were assessed in mice. In addition, F/HN-SIV was assessed in differentiated human air-liquid interface cultures, primary human nasal epithelial cells, and human and sheep lung slices. MEASUREMENTS AND MAIN RESULTS: A single dose produces lung expression for the lifetime of the mouse (~2 yr). Only brief contact time is needed to achieve transduction. Repeated daily administration leads to a dose-related increase in gene expression. Repeated monthly administration to mouse lower airways is feasible without loss of gene expression. There is no evidence of chronic toxicity during a 2-year study period. F/HN-SIV leads to persistent gene expression in human differentiated airway cultures and human lung slices and transduces freshly obtained primary human airway epithelial cells. CONCLUSIONS: The data support F/HN-pseudotyped SIV as a promising vector for pulmonary gene therapy for several diseases including CF. We are now undertaking the necessary refinements to progress this vector into clinical trials.


Asunto(s)
Fibrosis Quística/genética , Terapia Genética/métodos , Vectores Genéticos , Lentivirus/genética , Análisis de Varianza , Animales , Fibrosis Quística/terapia , Modelos Animales de Enfermedad , Femenino , Técnicas de Transferencia de Gen , Humanos , Pulmón/efectos de los fármacos , Pulmón/patología , Pulmón/fisiopatología , Ratones , Ratones Endogámicos C57BL , Virus de la Inmunodeficiencia de los Simios
4.
J Gene Med ; 14(7): 491-500, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22711445

RESUMEN

BACKGROUND: The nuclear membrane of differentiated airway epithelial cells is a significant barrier for nonviral vectors. Trans-cyclohexane-1,2-diol (TCHD) is an amphipathic alcohol that has been shown to collapse nuclear pore cores and allow the uptake of macromolecules that would otherwise be too large for nuclear entry. Previous studies have shown that TCHD can increase lipid-mediated transfection in vitro. METHODS: We aimed to reproduce these in vitro studies using the cationic lipid GL67A, which we are currently assessing in cystic fibrosis trials and, more importantly, we assessed the effects of TCHD on transfection efficiency in differentiated airway epithelium ex vivo and in mouse lung in vivo using three different drug delivery protocols (nebulisation and bolus administration of TCHD to the mouse lung, as well as perfusion of TCHD to the nasal epithelium, which prolongs contact time between the airway epithelium and drug). RESULTS: TCHD (0.5-2%) dose-dependently increased Lipofectamine 2000 and GL67A-mediated transfection of 293T cells by up to 2 logs. Encouragingly, exposure to 8% TCHD (but not 0.5% or 2.0%) increased gene expression in fully differentiated human air liquid interface cultures by approximately 20-fold, although this was accompanied by significant cell damage. However, none of the TCHD treated mice in any of the three protocols had higher gene expression compared to no TCHD controls. CONCLUSIONS: Although TCHD significantly increases gene transfer in cell lines and differentiated airway epithelium ex vivo, this effect is lost in vivo and further highlights that promising in vitro findings often cannot be translated into in vivo applications.


Asunto(s)
Ciclohexanos/farmacología , Ciclohexanoles/farmacología , Técnicas de Transferencia de Gen , Poro Nuclear/efectos de los fármacos , Sistema Respiratorio/efectos de los fármacos , Animales , Células Cultivadas , Ciclohexanos/administración & dosificación , Relación Dosis-Respuesta a Droga , Vías de Administración de Medicamentos , Epitelio/efectos de los fármacos , Femenino , Terapia Genética , Vectores Genéticos , Humanos , Lípidos/farmacología , Pulmón/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Mucosa Nasal/efectos de los fármacos , Transfección
5.
Mol Ther ; 18(6): 1173-82, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20332767

RESUMEN

Gene therapy for cystic fibrosis (CF) is making encouraging progress into clinical trials. However, further improvements in transduction efficiency are desired. To develop a novel gene transfer vector that is improved and truly effective for CF gene therapy, a simian immunodeficiency virus (SIV) was pseudotyped with envelope proteins from Sendai virus (SeV), which is known to efficiently transduce unconditioned airway epithelial cells from the apical side. This novel vector was evaluated in mice in vivo and in vitro directed toward CF gene therapy. Here, we show that (i) we can produce relevant titers of an SIV vector pseudotyped with SeV envelope proteins for in vivo use, (ii) this vector can transduce the respiratory epithelium of the murine nose in vivo at levels that may be relevant for clinical benefit in CF, (iii) this can be achieved in a single formulation, and without the need for preconditioning, (iv) expression can last for 15 months, (v) readministration is feasible, (vi) the vector can transduce human air-liquid interface (ALI) cultures, and (vii) functional CF transmembrane conductance regulator (CFTR) chloride channels can be generated in vitro. Our data suggest that this lentiviral vector may provide a step change in airway transduction efficiency relevant to a clinical programme of gene therapy for CF.


Asunto(s)
Fibrosis Quística/terapia , Terapia Genética , Vectores Genéticos , Lentivirus/genética , Virus Sendai/genética , Proteínas del Envoltorio Viral/genética , Animales , Diferenciación Celular , Línea Celular , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Transducción Genética
6.
Acta Biochim Biophys Sin (Shanghai) ; 42(1): 45-51, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20043046

RESUMEN

The aim of the study was to assess if low-frequency ultrasound (US), in the range of 30-35 kHz, increases non-viral gene transfer to the mouse lung. US is greatly attenuated in the lung due to large energy losses at the air/tissue interfaces. The advantages of low-frequency US, compared with high-frequency US are: (i) increased cavitation (responsible for the formation of transient pores in the cell membrane) and (ii) reduced energy losses during lung penetration. Cationic lipid GL67/plasmid DNA (pDNA), polyethylenimine (PEI)/pDNA and naked pDNA were delivered via intranasal instillation and the animals were then exposed to US (sonoporation) at 0.07 or 0.1 MPa for 10 min. Under these conditions, US did not enhance GL67 or PEI-mediated transfection. It did, however, increase naked pDNA gene transfer by approximately 4 folds. Importantly, this was achieved in the absence of microbubbles, which are crucial for the commonly used high-frequency (1 MHz) sonoporation but may not be able to withstand nebulization in a clinically relevant setup. Lung hemorrhage was also assessed and shown to increase with US pressure in a dose-dependent manner. We have thus, established that low-frequency US can enhance lung gene transfer with naked pDNA and this enhancement is more effective than the previously reported 1 MHz US.


Asunto(s)
Pulmón/virología , Polietileneimina/química , Transfección/métodos , Animales , Técnicas de Transferencia de Gen , Pulmón/química , Ratones , Transfección/estadística & datos numéricos , Ultrasonido
7.
J Gene Med ; 12(1): 55-63, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19937989

RESUMEN

BACKGROUND: When assessing the efficacy of gene transfer agents (GTAs) for cystic fibrosis (CF) gene therapy, we routinely evaluate gene transfer in the mouse nose and measure transfection efficiency by assessing transgene-specific mRNA using the real-time (TaqMan) quantitative reverse transcriptase-polymerase chain reaction. TaqMan is traditionally used to quantify expression in whole tissue homogenates, which in the nose would contain many cells types, including respiratory and olfactory epithelium. Only the respiratory epithelium is a satisfactory model for human airway epithelium and therefore CFTR gene transfer should be specifically assessed in respiratory epithelial cells (RECs). METHODS: We have compared laser microdissection, pronase digestion and nasal brushing for: (i) the ability to enrich RECs from the wild-type mouse nose and (ii) the length of time to perform the procedure. Using TaqMan, we subsequently assessed gene transfer in enriched RECs after nasal perfusion of GL67A/pCF1-CFTR complexes in a CF mouse model. RESULTS: Laser microdissection successfully isolated RECs; however, time-consuming sample preparation made this technique unsuitable for high-throughput studies. Pronase digestion was sufficiently rapid but only yielded 19% (range = 13%) RECs (n = 6). The nasal brushing method was superior, yielding 92% (range = 15%) RECs (n = 8) and was equally effective in CF knockout mice (91%, range = 14%, n = 10). Importantly, gene transfer was detectable in brushed RECs from 70% of perfused mice and the number of vector-specific transcripts was comparable to 3.5% of endogenous wild-type Cftr levels. CONCLUSIONS: Isolation of RECs by brushing allows accurate assessment of GTA transfection efficiency in an experimental system that is relevant for CF gene therapy.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Regulación de la Expresión Génica , Cavidad Nasal/patología , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/patología , Transgenes/genética , Animales , Separación Celular , Células Epiteliales/metabolismo , Células Epiteliales/patología , Proteínas de Unión a Ácidos Grasos/metabolismo , Técnicas de Transferencia de Gen , Vectores Genéticos/genética , Humanos , Rayos Láser , Ratones , Ratones Endogámicos C57BL , Microdisección , Cavidad Nasal/metabolismo , Tabique Nasal/metabolismo , Tabique Nasal/patología , Pronasa/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo
8.
Am J Respir Cell Mol Biol ; 43(1): 46-54, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19648474

RESUMEN

A clinical program to assess whether lipid GL67A-mediated gene transfer can ameliorate cystic fibrosis (CF) lung disease is currently being undertaken by the UK CF Gene Therapy Consortium. We have evaluated GL67A gene transfer to the murine nasal epithelium of wild-type and CF knockout mice to assess this tissue as a test site for gene transfer agents. The plasmids used were regulated by either (1) the commonly used short-acting cytomegalovirus promoter/enhancer or (2) the ubiquitin C promoter. In a study of approximately 400 mice with CF, vector-specific CF transmembrane conductance regulator (CFTR) mRNA was detected in nasal epithelial cells of 82% of mice treated with a cytomegalovirus-plasmid (pCF1-CFTR), and 62% of mice treated with an ubiquitin C-plasmid. We then assessed whether CFTR gene transfer corrected a panel of CFTR-specific endpoint assays in the murine nose, including ion transport, periciliary liquid height, and ex vivo bacterial adherence. Importantly, even with the comparatively large number of animals assessed, the CFTR function studies were only powered to detect changes of more than 50% toward wild-type values. Within this limitation, no significant correction of the CF phenotype was detected. At the current levels of gene transfer efficiency achievable with nonviral vectors, the murine nose is of limited value as a stepping stone to human trials.


Asunto(s)
Técnicas de Transferencia de Gen , Nariz/patología , Animales , Adhesión Bacteriana , Fibrosis Quística/genética , Citomegalovirus/genética , Elementos de Facilitación Genéticos , Femenino , Terapia Genética/métodos , Liposomas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Plásmidos/metabolismo , Regiones Promotoras Genéticas
9.
Biomaterials ; 31(9): 2665-72, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20022367

RESUMEN

We have assessed whether viscoelastic gels known to inhibit mucociliary clearance can increase lipid-mediated gene transfer. Methylcellulose or carboxymethylcellulose (0.25-1.5%) was mixed with complexes of the cationic lipid GL67A and plasmids encoding luciferase and perfused onto the nasal epithelium of mice. Survival after perfusion with 1% CMC or 1% MC was 90 and 100%, respectively. In contrast 1.5% CMC was uniformly lethal likely due to the viscous solution blocking the airways. Perfusion with 0.5% CMC containing lipid/DNA complexes reproducibly increased gene expression by approximately 3-fold (n=16, p<0.05). Given this benefit, likely related to increased duration of contact, we also assessed the effect of prolonging contact time of the liposome/DNA complexes by delivering our standard 80 microg DNA dose over either approximately 22 or 60 min of perfusion. This independently increased gene transfer by 6-fold (n=8, p<0.05) and could be further enhanced by the addition of 0.5% CMC, leading to an overall 25-fold enhancement (n=8, p<0.001) in gene expression. As a result of these interventions CFTR transgene mRNA transgene levels were increased several logs above background. Interestingly, this did not lead to correction of the ion transport defects in the nasal epithelium of cystic fibrosis mice nor for immunohistochemical quantification of CFTR expression. To assess if 0.5% CMC also increased gene transfer in the mouse lung, we used whole body nebulisation chambers. CMC was nebulised for 1h immediately before, or simultaneously with GL67A/pCIKLux. The former did not increase gene transfer, whereas co-administration significantly increased gene transfer by 4-fold (p<0.0001, n=18). This study suggests that contact time of non-viral gene transfer agents is a key factor for gene delivery, and suggests two methods which may be translatable for use in man.


Asunto(s)
Carboximetilcelulosa de Sodio/metabolismo , Técnicas de Transferencia de Gen , Sistema Respiratorio/metabolismo , Animales , Línea Celular , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Geles , Regulación de la Expresión Génica , Vectores Genéticos/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Potenciales de la Membrana , Ratones , Nebulizadores y Vaporizadores , Perfusión , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Factores de Tiempo , Virus/genética
10.
Biomaterials ; 30(10): 1971-7, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19152975

RESUMEN

Proteasome inhibitors have been shown to increase adeno-associated virus (AAV)-mediated transduction in vitro and in vivo. To assess if proteasome inhibitors also increase lipid-mediated gene transfer with relevance to cystic fibrosis (CF), we first assessed the effects of doxorubicin and N-acetyl-l-leucinyl-l-leucinal-l-norleucinal in non-CF (A549) and CF (CFTE29o-) airway epithelial cell lines. CFTE29o- cells did not show a response to Dox or LLnL; however, gene transfer in A549 cells increased in a dose-related fashion (p < 0.05), up to approximately 20-fold respectively at the optimal dose (no treatment: 9.3 x 10(4) +/- 1.5 x 10(3), Dox: 1.6 x 10(6)+/-2.6 x 10(5), LLnL: 1.9 x 10(6) +/- 3.2 x 10(5)RLU/mg protein). As Dox is used clinically in cancer chemotherapy we next assessed the effect of this drug on non-viral lung gene transfer in vivo. CF knockout mice were injected intraperitoneally (IP) with Dox (25-100 mg/kg) immediately before nebulisation with plasmid DNA carrying a luciferase reporter gene under the control of a CMV promoter/enhancer (pCIKLux) complexed to the cationic lipid GL67A. Dox also significantly (p < 0.05) increased expression of a plasmid regulated by an elongation factor 1alpha promoter (hCEFI) approximately 8-fold. Although administration of Dox before lung gene transfer may not be a clinically viable option, understanding how Dox increases lung gene expression may help to shed light on intracellular bottle-necks to gene transfer, and may help to identify other adjuncts that may be more appropriate for use in man.


Asunto(s)
Fibrosis Quística/genética , Fibrosis Quística/terapia , Doxorrubicina/farmacología , Técnicas de Transferencia de Gen , Vectores Genéticos/administración & dosificación , Pulmón/efectos de los fármacos , Animales , Línea Celular , Femenino , Vectores Genéticos/química , Humanos , Pulmón/metabolismo , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 1 de Elongación Peptídica/genética , Regiones Promotoras Genéticas/genética
11.
Am J Respir Cell Mol Biol ; 39(4): 490-6, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18458238

RESUMEN

Attempts at correcting the nasal potential difference (PD) in cystic fibrosis (CF) mice have long been used in preclinical gene and small molecule therapy development. However, in general, CF mice suffer from intestinal disease, are runted, and have high mortality rates; they are therefore difficult to work with, especially if large numbers are required. Because of this, large-scale PD studies in CF mice have not been performed. Working with CF mice has become substantially easier after the generation of the gut-corrected CF-knockout mouse. Fatty acid-binding promoter (FABp)-mediated expression of CFTR in the gut, but not the airways, prevents the intestinal disease of the CF knockout mouse. This model has given us the unique opportunity to systematically study PDs in large numbers of CF mice. The nose, but not the lungs, of these animals mimic the bioelectric defect seen in humans. We have therefore assessed the bioelectrics of the respiratory epithelium comparing FABp-CF and wild-type mice. The large body of data gathered in CF and wild-type mice allowed us, for the first time, to establish power calculations that should inform sample sizes required in gene and small molecule therapy development. In addition, we address the important issues of intra-animal variability as well as intra- and inter-operator variability for scoring the traces, and the effect of age and sex on nasal PD in CF mice. These data should allow a more informed use of CF animals in future studies.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/biosíntesis , Fibrosis Quística/fisiopatología , Mucosa Nasal/fisiopatología , Animales , Fibrosis Quística/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Proteínas de Unión a Ácidos Grasos/genética , Femenino , Mucosa Intestinal/metabolismo , Masculino , Potenciales de la Membrana , Ratones , Ratones Noqueados , Mucosa Nasal/metabolismo , Regiones Promotoras Genéticas
12.
Biomaterials ; 29(10): 1533-40, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18155138

RESUMEN

Imaging of in vivo gene expression using luciferase expression in various organs has been used for several years. In contrast to other organs, in vivo imaging of the lung, particularly after non-viral gene transfer has not been extensively studied. The aim of this study was to address several questions: (1) Does in vivo light emission correlate with standard tissue homogenate-based luciferase detection in a dose-dependent manner? Recombinant Sendai virus (SeV) transduces airway epithelial cells very efficiently and was used to address this question, (2) Is the sensitivity of the assay sufficient to detect non-viral gene transfer? We treated mice with SeV-Lux vector using our standard "sniffing" protocol, a method that predominantly results in lung deposition. Dose-related in vivo light emission was visible in all animals. Importantly, there was a significant correlation (r>0.90, p<0.0001) between the in vivo and ex vivo assays in both the left and right lung. We next transfected the nasal epithelium via nasal perfusion or the lungs ("sniffing") of mice with a luciferase plasmid (pCIKLux) complexed to the cationic lipid GL67 (n=25-27/group) and imaged luciferase expression in vivo 24h after transfection. Gene expression was detectable in both organs. Correlation between the in vivo and ex vivo assays was significant (r=0.52, p<0.005) in the left, but not the right lung. The correlation in the nose was weaker (r=0.45, p<0.05). To our knowledge these studies show for the first time that this non-invasive method of assessing pulmonary gene transfer is viable for evaluating non-viral gene transfer agents.


Asunto(s)
Mediciones Luminiscentes/métodos , Sistema Respiratorio/metabolismo , Transfección/métodos , Animales , Femenino , Vectores Genéticos/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Luciferasas/genética , Luciferasas/metabolismo , Pulmón/metabolismo , Ratones , Ratones Endogámicos BALB C , Mucosa Nasal/metabolismo , Virus Sendai/genética
13.
Respir Res ; 7: 26, 2006 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-16480492

RESUMEN

BACKGROUND: The cationic lipid Genzyme lipid (GL) 67 is the current "gold-standard" for in vivo lung gene transfer. Here, we assessed, if GL67 mediated uptake of siRNAs and asODNs into airway epithelium in vivo. METHODS: Anti-lacZ and ENaC (epithelial sodium channel) siRNA and asODN were complexed to GL67 and administered to the mouse airway epithelium in vivo Transfection efficiency and efficacy were assessed using real-time RT-PCR as well as through protein expression and functional studies. In parallel in vitro experiments were carried out to select the most efficient oligonucleotides. RESULTS: In vitro, GL67 efficiently complexed asODNs and siRNAs, and both were stable in exhaled breath condensate. Importantly, during in vitro selection of functional siRNA and asODN we noted that asODNs accumulated rapidly in the nuclei of transfected cells, whereas siRNAs remained in the cytoplasm, a pattern consistent with their presumed site of action. Following in vivo lung transfection siRNAs were only visible in alveolar macrophages, whereas asODN also transfected alveolar epithelial cells, but no significant uptake into conducting airway epithelial cells was seen. SiRNAs and asODNs targeted to beta-galactosidase reduced betagal mRNA levels in the airway epithelium of K18-lacZ mice by 30% and 60%, respectively. However, this was insufficient to reduce protein expression. In an attempt to increase transfection efficiency of the airway epithelium, we increased contact time of siRNA and asODN using the in vivo mouse nose model. Although highly variable and inefficient, transfection of airway epithelium with asODN, but not siRNA, was now seen. As asODNs more effectively transfected nasal airway epithelial cells, we assessed the effect of asODN against ENaC, a potential therapeutic target in cystic fibrosis; no decrease in ENaC mRNA levels or function was detected. CONCLUSION: This study suggests that although siRNAs and asODNs can be developed to inhibit gene expression in culture systems and certain organs in vivo, barriers to nucleic acid transfer in airway epithelial cells seen with large DNA molecules may also affect the efficiency of in vivo uptake of small nucleic acid molecules.


Asunto(s)
Fibrosis Quística/genética , Fibrosis Quística/metabolismo , Marcación de Gen/métodos , Lípidos/química , Oligonucleótidos Antisentido/genética , ARN Interferente Pequeño/genética , Transfección/métodos , Animales , Células Cultivadas , Células Epiteliales , Silenciador del Gen , Humanos , Ratones , Células 3T3 NIH , Oligonucleótidos Antisentido/administración & dosificación , ARN Interferente Pequeño/administración & dosificación , Mucosa Respiratoria
14.
Am J Respir Crit Care Med ; 171(9): 1015-9, 2005 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-15640364

RESUMEN

Nasal potential difference measurements are valuable endpoint assays in clinical studies of novel treatments for cystic fibrosis (CF). Similar measurements made on the lower airway via the bronchoscope have been successful in adults, but have not been reported in children, the group most likely to benefit from such therapies. Here we report the design and validation of a small, single-lumen catheter technique allowing baseline potential difference and chloride secretion to be assessed in the distal airways of children as young as 1 year of age. Tracheal baseline values were significantly higher in children with CF than those without, although this was not the case more distally. In airways between the third and seventh generation, perfusion with a zero chloride solution containing isoprenaline led to a significant change in potential difference in children without CF, whereas no change was seen in those with CF. This measure provided a reliable distinguishing test between the two disease groups. We confirm that invasive bronchoscopic techniques can be performed safely and reliably in small children. Potential difference measurements could form a useful functional endpoint assay for future studies of either the CFTR gene or protein-based therapies in future trials in the pediatric age group.


Asunto(s)
Bronquios/fisiología , Bronquios/fisiopatología , Cateterismo/métodos , Cloruros/análisis , Fibrosis Quística/fisiopatología , Potenciales de la Membrana , Tráquea/fisiología , Tráquea/fisiopatología , Broncoscopía , Niño , Preescolar , Humanos , Lactante , Transporte Iónico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...