Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
1.
Nat Genet ; 25(2): 187-91, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10835634

RESUMO

There is much variability between individuals in the response to inhaled toxins, but it is not known why certain people develop disease when challenged with environmental agents and others remain healthy. To address this, we investigated whether TLR4 (encoding the toll-like receptor-4), which has been shown to affect lipopolysaccharide (LPS) responsiveness in mice, underlies the variability in airway responsiveness to inhaled LPS in humans. Here we show that common, co-segregating missense mutations (Asp299Gly and Thr399Ile) affecting the extracellular domain of the TLR4 receptor are associated with a blunted response to inhaled LPS in humans. Transfection of THP-1 cells demonstrates that the Asp299Gly mutation (but not the Thr399Ile mutation) interrupts TLR4-mediated LPS signalling. Moreover, the wild-type allele of TLR4 rescues the LPS hyporesponsive phenotype in either primary airway epithelial cells or alveolar macrophages obtained from individuals with the TLR4 mutations. Our findings provide the first genetic evidence that common mutations in TLR4 are associated with differences in LPS responsiveness in humans, and demonstrate that gene-sequence changes can alter the ability of the host to respond to environmental stress.


Assuntos
Proteínas de Drosophila , Lipopolissacarídeos/farmacologia , Macrófagos Alveolares/fisiologia , Glicoproteínas de Membrana/genética , Mutação de Sentido Incorreto/genética , Receptores de Superfície Celular/genética , Mucosa Respiratória/fisiologia , Administração por Inalação , Adolescente , Adulto , Alelos , Sequência de Aminoácidos , Sequência de Bases , Células Cultivadas , Análise Mutacional de DNA , Feminino , Volume Expiratório Forçado/efeitos dos fármacos , Humanos , Lipopolissacarídeos/administração & dosagem , Macrófagos Alveolares/efeitos dos fármacos , Masculino , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/metabolismo , Pessoa de Meia-Idade , Dados de Sequência Molecular , Receptores de Superfície Celular/química , Receptores de Superfície Celular/metabolismo , Hipersensibilidade Respiratória/induzido quimicamente , Hipersensibilidade Respiratória/genética , Hipersensibilidade Respiratória/fisiopatologia , Mucosa Respiratória/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Receptor 4 Toll-Like , Receptores Toll-Like
2.
Nat Genet ; 6(1): 75-83, 1994 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-7511023

RESUMO

Gene therapy for cystic fibrosis (CF) will require the safe transfer of CFTR cDNA to airway epithelia in vivo. We showed previously that a recombinant adenovirus, Ad2/CFTR-1, expresses CFTR in vitro. As adenovirus rarely integrates, treatment will require repeated vector administration. We applied Ad2/CFTR-1 to intrapulmonary airway epithelia of cotton rats and nasal epithelia of Rhesus monkeys. In both species we detected CFTR mRNA and protein after repeated administration and in monkeys, protein was detected six weeks after repeat administration. The vector did not replicate and was rapidly cleared. Despite an antibody response, there was no evidence of a local or systemic inflammatory response after repeat administration. These data indicate that repetitive administration of Ad2/CFTR-1 is both safe and efficacious.


Assuntos
DNA Complementar/administração & dosagem , DNA Complementar/genética , Terapia Genética/métodos , Proteínas de Membrana/genética , Adenoviridae/genética , Animais , Sequência de Bases , Fibrose Cística/genética , Fibrose Cística/terapia , Regulador de Condutância Transmembrana em Fibrose Cística , Primers do DNA/genética , Epitélio/metabolismo , Feminino , Expressão Gênica , Terapia Genética/efeitos adversos , Vetores Genéticos , Humanos , Macaca mulatta , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , Sistema Respiratório/metabolismo , Sistema Respiratório/patologia , Segurança , Sigmodontinae
3.
Adv Exp Med Biol ; 660: 183-93, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20221881

RESUMO

Pseudomonas aeruginosa is a Gram-negative bacterium which exacts a heavy burden on immunocompromised patients, but is non-pathogenic in a healthy host. Using small signaling molecules called acyl-homoserine lactones (AHLs), populations of P. aeruginosa can coordinate phenotypic changes, including biofilm formation and virulence factor secretion. This concentration-dependent process is called quorum sensing (QS). Interference with QS has been identified as a potential source of new treatments for P. aeruginosa infection. The human enzyme paraoxonase 1 (PON1) degrades AHL molecules, and is a promising candidate for QS interference therapy. Although paraoxonase orthologs exist in many species, genetic redundancy in humans and other mammals has made studying the specific effects of PON1 quite difficult. Arthropods, however, do not express any PON homologs. We generated a novel model to study the specific effects of PON1 by transgenically expressing human PON1 in Drosophila melanogaster. Using this model, we showed that P. aeruginosa infection lethality is QS-dependent, and that expression of PON1 has a protective effect. This work demonstrates the value of a D. melanogaster model for investigating the specific functions of members of the paraoxonase family in vivo, and suggests that PON1 plays a role in innate immunity.


Assuntos
Arildialquilfosfatase/metabolismo , Infecções por Pseudomonas/metabolismo , Pseudomonas aeruginosa/metabolismo , Sequência de Aminoácidos , Animais , Biofilmes , Drosophila melanogaster , Humanos , Imunidade Inata , Modelos Biológicos , Modelos Químicos , Dados de Sequência Molecular , Filogenia , Percepção de Quorum , Homologia de Sequência de Aminoácidos , Transgenes , Fatores de Virulência
4.
J Clin Invest ; 100(5): 1144-9, 1997 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-9276731

RESUMO

Although recombinant adenoviruses are attractive vectors for gene transfer to airway epithelia, they have proven to be relatively inefficient. To investigate the mechanisms of adenovirus-mediated gene transfer to airway epithelia, we examined the role of adenovirus fiber and penton base, the two proteins involved in attachment to and entry of virus into the cell. We used human airway epithelia grown under conditions that allow differentiation and development of a ciliated apical surface that closely resembles the in vivo condition. We found that addition of fiber protein inhibited virus binding and vector-mediated gene transfer to immature airway epithelia, as well as to primary cultures of rat hepatocytes and HeLa cells. However, fiber protein had no effect on vector binding and gene transfer to ciliated airway epithelia. We obtained similar results with addition of penton base protein: the protein inhibited gene transfer to immature epithelia, whereas there was no effect with ciliated epithelia. Moreover, infection was not attenuated with an adenovirus containing a mutation in penton base that prevents the interaction with cell surface integrins. These data suggest that the receptors required for efficient infection by adenovirus are either not present or not available on the apical surface of ciliated human airway epithelia. The results explain the reason for inefficient gene transfer and suggest approaches for improvement.


Assuntos
Adenoviridae/genética , Brônquios/virologia , Proteínas do Capsídeo , Capsídeo/fisiologia , Técnicas de Transferência de Genes , Receptores Virais/fisiologia , Traqueia/virologia , Animais , Células Cultivadas , Epitélio/virologia , Células HeLa , Humanos , Ratos
5.
J Clin Invest ; 102(1): 184-93, 1998 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-9649572

RESUMO

Adenovirus (Ad)-mediated gene transfer to airway epithelia is inefficient because the apical membrane lacks the receptor activity to bind adenovirus fiber protein. Calcium phosphate (CaPi) precipitates have been used to deliver plasmid DNA to cultured cell lines. However, such precipitates are not effective in many primary cultures or in vivo. Here we show that incorporating recombinant adenovirus into a CaPi coprecipitate markedly enhances transgene expression in cells that are resistant to adenovirus infection. Enhancement requires that the virus be contained in the precipitate and viral proteins are required to increase expression. Ad: CaPi coprecipitates increase gene transfer by increasing fiber-independent binding of virus to cells. With differentiated cystic fibrosis (CF) airway epithelia in vitro, a 20-min application of Ad:CaPi coprecipitates that encode CF transmembrane conductance regulator produced as much CF transmembrane conductance regulator Cl- current as a 24-h application of adenovirus alone. We found that Ad:CaPi coprecipitates also increased transgene expression in mouse lung in vivo; importantly, expression was particularly prominent in airway epithelia. These results suggest a new mechanism for gene transfer that may be applicable to a number of different gene transfer applications and could be of value in gene transfer to CF airway epithelia in vivo.


Assuntos
Adenoviridae/genética , Fosfatos de Cálcio/administração & dosagem , Proteínas do Capsídeo , Fibrose Cística/terapia , Técnicas de Transferência de Genes , Vetores Genéticos , Pulmão/virologia , Células 3T3 , Animais , Células COS , Capsídeo/fisiologia , Precipitação Química , Fibrose Cística/genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL
6.
J Clin Invest ; 97(6): 1504-11, 1996 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-8617884

RESUMO

Cystic fibrosis (CF) is a common autosomal recessive disease caused by mutations in the CF transmembrane conductance regulator gene. Recombinant adenoviruses have shown promise as vectors for transfer of CF transmembrane conductance regulator cDNA to airway epithelia and correction of the Cl- transport defect. However, because adenovirus-mediated gene transfer is transient, use of adenovirus as a vector for treatment of CF would require repeated administration. Therefore, we evaluated repeat administration of an adenovirus vector to the nasal epithelium of patients with CF with five escalating doses of up to 10(10) infectious units. There were no detectable adverse affects. All subjects were initially seropositive but developed additional humoral immune responses. The vector partially corrected the defect in airway epithelial Cl- transport in some subjects, although there was variability between subjects and there was less correction with subsequent administration, perhaps because the immune response limited gene transfer. Future work must focus on vectors with increased efficiency and with the ability to evade host defenses.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/uso terapêutico , Fibrose Cística/terapia , Mucosa Nasal/metabolismo , Adenoviridae , Adulto , Fibrose Cística/genética , Fibrose Cística/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Sistemas de Liberação de Medicamentos , Epitélio/metabolismo , Feminino , Técnicas de Transferência de Genes , Terapia Genética , Vetores Genéticos , Humanos , Masculino , Pessoa de Meia-Idade
7.
J Clin Invest ; 100(6): 1529-37, 1997 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-9294121

RESUMO

Cationic lipids show promise as vectors for transfer of CFTR cDNA to airway epithelia of patients with cystic fibrosis (CF). However, previous studies have not compared the effect of DNA-lipid to DNA alone. Recently, we developed a formulation of plasmid encoding CFTR (pCF1-CFTR) and cationic lipid (GL-67:DOPE) that generated greater gene transfer in mouse lung than previously described DNA-lipid vectors. Therefore, we tested the hypothesis that DNA-lipid complexes were more effective than DNA alone at transferring CFTR cDNA to airway epithelia in vivo. We administered complexes of DNA-lipid to one nostril and DNA alone to the other nostril in a randomized, double-blind study. Electrophysiologic measurements showed that DNA-lipid complexes partially corrected the Cl- transport defect. Importantly, the pCF1-CFTR plasmid alone was at least as effective as complexes of DNA with lipid. Measurements of vector-specific CFTR transcripts also showed gene transfer with both DNA-lipid and DNA alone. These results indicate that nonviral vectors can transfer CFTR cDNA to airway epithelia and at least partially restore the Cl- transport defect characteristic of CF. However, improvements in the overall efficacy of gene transfer are required to develop a treatment for CF.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/genética , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Fibrose Cística/terapia , DNA/administração & dosagem , Técnicas de Transferência de Genes , Mucosa Nasal/metabolismo , Adolescente , Adulto , Amilorida/farmacologia , Cloro/farmacologia , Fibrose Cística/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/imunologia , DNA/imunologia , DNA/farmacocinética , Relação Dose-Resposta a Droga , Método Duplo-Cego , Estudos de Avaliação como Assunto , Feminino , Vetores Genéticos , Humanos , Interleucina-6/sangue , Lipídeos/imunologia , Lipídeos/farmacocinética , Masculino , Potenciais da Membrana/efeitos dos fármacos , Pessoa de Meia-Idade , Mucosa Nasal/efeitos dos fármacos , Mucosa Nasal/fisiologia , Reação em Cadeia da Polimerase , Terbutalina/farmacologia , Fatores de Tempo , Resultado do Tratamento
8.
J Clin Invest ; 102(7): 1421-30, 1998 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-9769335

RESUMO

Previous studies have suggested that alveolar Na,K-ATPases play an important role in active Na+ transport and lung edema clearance. We reasoned that overexpression of Na,K-ATPase subunit genes could increase Na,K-ATPase function in lung epithelial cells and edema clearance in rat lungs. To test this hypothesis we produced replication deficient human type 5 adenoviruses containing cDNAs for the rat alpha1 and beta1 Na,K-ATPase subunits (adMRCMValpha1 and adMRCMVbeta1, respectively). As compared to controls, adMRCMVbeta1 increased beta1 subunit expression and Na,K-ATPase function by 2. 5-fold in alveolar type 2 epithelial cells and rat airway epithelial cell monolayers. No change in Na,K-ATPase function was noted after infection with adMRCMValpha1. Rat lungs infected with adMRCMVbeta1, but not adMRCMValpha1, had increased beta1 protein levels and lung liquid clearance 7 d after tracheal instillation. Alveolar epithelial permeability to Na+ and mannitol was mildly increased in animals infected with adMRCMVbeta1 and a similar Escherichia coli lacZ-expressing virus. Our data shows, for the first time, that transfer of the beta1 Na,K-ATPase subunit gene augments Na,K-ATPase function in epithelial cells and liquid clearance in rat lungs. Conceivably, overexpression of Na,K-ATPases could be used as a strategy to augment lung liquid clearance in patients with pulmonary edema.


Assuntos
Células Epiteliais/fisiologia , Pulmão/fisiologia , Alvéolos Pulmonares/fisiologia , Edema Pulmonar/terapia , ATPase Trocadora de Sódio-Potássio/genética , Adenovírus Humanos , Animais , Permeabilidade da Membrana Celular , Células Cultivadas , Células Epiteliais/citologia , Vetores Genéticos , Humanos , Pulmão/citologia , Substâncias Macromoleculares , Masculino , Alvéolos Pulmonares/citologia , Alvéolos Pulmonares/enzimologia , Surfactantes Pulmonares/análise , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/biossíntese , Sódio/metabolismo , ATPase Trocadora de Sódio-Potássio/biossíntese
9.
J Clin Invest ; 96(4): 2051-64, 1995 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-7560099

RESUMO

The most common cause of cystic fibrosis is a mutation that deletes phenylalanine 508 in cystic fibrosis transmembrane conductance regulator (CFTR). The delta F508 protein is misprocessed and degraded rather than traveling to the apical membrane. We used a novel strategy to introduce the delta F508 mutation into the mouse CFTR gene. Affected epithelia from homozygous delta F508 mice lacked CFTR in the apical membrane and were Cl-impermeable. These abnormalities are the same as those observed in patients with delta F508 and suggest that these mice have the same cellular defect. 40% of homozygous delta F508 animals survived into adulthood and displayed several abnormalities found in human disease and in CFTR null mice. These animals should provide an excellent model to investigate pathogenesis and to examine therapies directed at correcting the delta F508 defect.


Assuntos
Alelos , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Fibrose Cística/genética , Animais , Sequência de Bases , Fibrose Cística/patologia , Sistema Digestório/metabolismo , Sistema Digestório/patologia , Modelos Animais de Doenças , Eletrólitos/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Mutação , Ductos Pancreáticos/metabolismo , RNA Mensageiro/análise
10.
J Clin Invest ; 95(6): 2620-32, 1995 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-7539457

RESUMO

Vector-mediated gene transfer offers a direct method of correcting genetic pulmonary diseases and might also be used to correct temporary abnormalities associated with acquired, nongenetic disorders. Because the fetus or newborn may be a more immune tolerant host for gene transfer using viral vectors, we used replication defective recombinant adenoviral vectors to test the feasibility of gene transfer to the fetal pulmonary epithelium in vitro and in vivo. Both proximal and distal epithelial cells in cultured fetal lung tissues from rodents and humans diffusely expressed the lacZ transgene 3 d after viral infection. In vivo gene delivery experiments were performed in fetal mice and lambs. Delivery of Ad2/CMV-beta Gal to the amniotic fluid in mice produced intense transgene expression in the fetal epidermis and amniotic membranes, some gastrointestinal expression, but no significant airway epithelial expression. When we introduced the adenoviral vector directly into the trachea of fetal lambs, the lacZ gene was expressed in the tracheal, bronchial, and distal pulmonary epithelial cells 3 d after viral infection. Unexpectedly, reactive hyperplasia and squamous metaplasia were noted in epithelia expressing lacZ in the trachea, but not in the distal lung of fetal lambs. 1 wk after infection, adenovirus-treated fetuses developed inflammatory cell infiltrates in the lung tissue with CD4, CD8, IgM, and granulocyte/macrophage positive immune effector cells. Transgene expression faded coincident with inflammation and serologic evidence of antiadenoviral antibody production. While these studies document the feasibility of viral-mediated gene transfer in the prenatal lung, they indicate that immunologic responses to E1-deleted recombinant adenoviruses limit the duration of transgene expression.


Assuntos
Adenovírus Humanos/genética , Técnicas de Transferência de Genes , Pulmão , Animais , Sequência de Bases , Regulador de Condutância Transmembrana em Fibrose Cística , Primers do DNA/química , Epitélio , Expressão Gênica , Vetores Genéticos , Células HeLa , Humanos , Técnicas In Vitro , Pulmão/embriologia , Proteínas de Membrana/genética , Camundongos , Dados de Sequência Molecular , RNA Mensageiro/genética , Ovinos , Fatores de Tempo , Traqueia/metabolismo
11.
J Clin Invest ; 105(5): 589-96, 2000 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10712430

RESUMO

Developing gene therapy for cystic fibrosis has been hindered by limited binding and endocytosis of vectors by human airway epithelia. Here we show that the apical membrane of airway epithelia express the urokinase plasminogen activator receptor (uPAR). Urokinase plasminogen activator (uPA), or a 7-residue peptide derived from this protein (u7-peptide), bound the receptor and stimulated apical endocytosis. Both ligands enhanced gene transfer by nonspecifically bound adenovirus and adeno-associated virus vectors and by a modified adenovirus vector that had been coupled to the u7-peptide. These data provide the first evidence that targeting an apical receptor can circumvent the two most important barriers to gene transfer in airway epithelia. Thus, the uPA/uPAR system may offer significant advantages for delivering genes and other pharmaceuticals to airway epithelia.


Assuntos
Técnicas de Transferência de Genes , Pulmão/metabolismo , Receptores de Superfície Celular/efeitos dos fármacos , Adenoviridae/genética , Adenoviridae/metabolismo , Animais , Células Cultivadas , Fibrose Cística/terapia , Endocitose , Citometria de Fluxo , Imunofluorescência , Terapia Genética/métodos , Haplorrinos , Humanos , Fragmentos de Peptídeos/farmacologia , Ligação Proteica , Receptores de Ativador de Plasminogênio Tipo Uroquinase , Ativador de Plasminogênio Tipo Uroquinase/farmacologia
12.
J Clin Invest ; 104(11): R55-62, 1999 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-10587528

RESUMO

Several problems limit the application of gene transfer to correct the cystic fibrosis (CF) Cl(-) transport defect in airway epithelia. These include inefficient transduction with vectors applied to the apical surface, a low rate of division by airway epithelial cells, failure of transgene expression to persist, and immune responses to vectors or vector-encoded proteins. To address these issues, we used a feline immunodeficiency virus-based (FIV-based) vector. FIV vector formulated with a calcium chelator transduced fully differentiated, nondividing human airway epithelia when applied to the apical surface. FIV-based vector encoding the cystic fibrosis transmembrane conductance regulator cDNA corrected the Cl(-) transport defect in differentiated CF airway epithelia for the life of the culture (>3 months). When this approach was applied in vivo, FIV vector expressing beta-galactosidase transduced 1-14% of adult rabbit airway epithelia. Transduced cells were present in the conducting airways, bronchioles, and alveoli. Importantly, gene expression persisted, and cells with progenitor capacity were targeted. FIV-based lentiviral vectors may be useful for the treatment of genetic lung diseases such as CF. This article may have been published online in advance of the print edition.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/genética , Terapia Genética/métodos , Vírus da Imunodeficiência Felina/genética , Pulmão/patologia , Animais , Cloretos/metabolismo , Fibrose Cística/terapia , DNA Complementar/genética , Células Epiteliais , Técnicas de Transferência de Genes , Vetores Genéticos , Humanos , Fatores de Tempo , Traqueia/metabolismo , Transdução Genética , beta-Galactosidase/genética
13.
Hum Gene Ther ; 10(4): 603-13, 1999 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-10094203

RESUMO

Gene transfer could provide a novel treatment for cystic fibrosis. However, current vectors, including recombinant adenoviruses, are relatively inefficient at gene transfer to airway epithelia. We have found that delivering adenovirus in a calcium phosphate coprecipitate (Ad:CaPi coprecipitates) enhanced the efficiency of gene transfer to airway epithelia in vitro and in vivo. However, the potential for injury to the epithelium was not evaluated. In NIH 3T3 cells treated with Ad:CaPi coprecipitates, we found that a 30-min exposure, which was sufficient for maximal transgene expression, produced no toxicity; whereas some other transfection reagents induced significant toxicity. Moreover, when Ad:CaPi coprecipitates were applied to the apical surface of differentiated airway epithelia in vitro, they did not reduce transepithelial resistance, even after prolonged incubation. Delivery of Ad:CaPi coprecipitates to mouse lung induced an inflammatory response, but it was not substantially different from that following administration of adenovirus alone. Thus, Ad:CaPi coprecipitates significantly enhance gene transfer to differentiated human airway epithelia in vitro and to mouse lung in vivo without increasing toxicity or the inflammatory response. Thus, CaPi coprecipitates may enhance the therapeutic index of adenovirus-based gene transfer vectors.


Assuntos
Adenoviridae/genética , Fosfatos de Cálcio/administração & dosagem , Técnicas de Transferência de Genes/normas , Vetores Genéticos , Traqueia/metabolismo , Células 3T3 , Animais , Líquido da Lavagem Broncoalveolar , Fosfatos de Cálcio/efeitos adversos , Células Cultivadas , Precipitação Química , Células Epiteliais/metabolismo , Terapia Genética/efeitos adversos , Pulmão/metabolismo , Pulmão/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL
14.
Hum Gene Ther ; 5(5): 585-93, 1994 May.
Artigo em Inglês | MEDLINE | ID: mdl-7519884

RESUMO

Adenovirus vectors are a promising vehicle to deliver cystic fibrosis transmembrane conductance regulator (CFTR) cDNA to airway epithelia. However, the value of adenovirus vectors will depend on the efficiency with which the vector can correct the defective fluid transport that is though to underlie the pathogenesis of the disease. To address the efficiency of gene transfer, we applied adenovirus vectors expressing CFTR (Ad2/CFTR-1) or beta-galactosidase to the mucosal surface of primary cultures of airway epithelial cells grown as polarized epithelial monolayers on permeable filter supports. These conditions provide a model that reproduces the physiology of the airways in vivo. We found that after adding 1 moi Ad2/CFTR-1 to the mucosal surface, cAMP agonists stimulated fluid secretion that was within the range observed in epithelia from normal subjects. When we measured electrolyte transport, we found that as little as 0.1 moi partially restored cAMP-stimulated Cl- secretion, and at 10 moi Cl- secretion was in the normal range. A related vector encoding beta-galactosidase generated activity in approximately 20% of cells at an moi of 1 and 90% of cells at an moi of 10. These data suggest that Ad2/CFTR-1 is very efficient at restoring normal fluid and electrolyte transport to CF airway epithelia. Thus, they suggest that relatively low input doses could be used for gene transfer to CF airway epithelia.


Assuntos
Adenoviridae/genética , Cloretos/metabolismo , AMP Cíclico/fisiologia , Fibrose Cística/metabolismo , Técnicas de Transferência de Genes , Vetores Genéticos , Pulmão/metabolismo , Proteínas de Membrana/metabolismo , Fibrose Cística/genética , Fibrose Cística/terapia , Regulador de Condutância Transmembrana em Fibrose Cística , Epitélio/metabolismo , Humanos , Proteínas de Membrana/genética , beta-Galactosidase/metabolismo
15.
Hum Gene Ther ; 10(16): 2615-26, 1999 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-10566889

RESUMO

Recombinant adenoviruses are currently being used as vectors for gene delivery to a wide variety of cells and tissues. Although generally efficacious for gene transfer in vitro, improvement in the efficiency of vector delivery in vivo may aid several gene therapy applications. One major obstacle is the lack of high-affinity viral receptors on the surface of certain cells that are targets for gene therapy. In principle, incorporation of avid, cell-specific ligands into the virion could markedly improve vector entry into the desired tissues. We have developed a strategy for addressing this issue in the lung by biopanning differentiated, ciliated airway epithelial cells against a phage display library. The peptide with the most effective binding was coupled to the surface of an adenovirus using bifunctional polyethylene glycol (PEG) molecules. The chemically modified adenoviral vector was able to effect gene transfer to well-differentiated human airway epithelial cells by an alternative pathway dependent on the incorporated peptide. Coupling of PEG to the surface of the virus also served to partially protect the virus from neutralizing antibodies in vitro. These experiments will aid in the design of improved adenoviral vectors with the capacity for more specific and efficient delivery of therapeutic genes to desired target tissues. We have used a novel method for enhancing gene delivery to target cells by coupling a biologically selected peptide to the surface of an adenovirus with bifunctional PEG molecules. Modification of the viral capsid by the addition of a peptide with binding preference for differentiated ciliated airway epithelia allowed gene delivery to those cells by a novel entry pathway. Incorporation of the CFTR gene in a similarly modified vector resulted in correction of defective Cl- transport in well-differentiated epithelial cultures established from human cystic fibrosis (CF) donors. The presence of PEG molecules on the surface of the virus served, in addition, to reduce antibody neutralization. Modification of adenoviruses with PEG/peptide complexes can serve to partially overcome the barrier of inefficient gene transfer in some cell types and some of the adverse immunological responses associated with gene delivery by these vectors.


Assuntos
Adenoviridae/genética , Técnicas de Transferência de Genes , Vetores Genéticos/química , Peptídeos/metabolismo , Peptídeos/farmacologia , Adenoviridae/química , Adenoviridae/imunologia , Anticorpos/metabolismo , Bacteriófagos , Brônquios/citologia , Brônquios/metabolismo , Células Cultivadas , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus , Sistemas de Liberação de Medicamentos , Células Epiteliais/metabolismo , Vetores Genéticos/metabolismo , Humanos , Soros Imunes , Testes de Neutralização , Biblioteca de Peptídeos , Peptídeos/química , Polietilenoglicóis/química , Receptores Virais/metabolismo , Transdução Genética
16.
Hum Gene Ther ; 8(9): 1087-93, 1997 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-9189766

RESUMO

Gene transfer with integrating vectors such as recombinant retrovirus has the potential to correct inherited lung diseases permanently. As a gene therapy target, the pulmonary epithelium presents several challenges to vector delivery in vivo. Many of the host defenses that have evolved to prevent infection from inhaled bacteria or viruses represent potential barriers to gene transfer to the lung. We performed in vitro studies to determine whether two components of the innate immune system of the lung, airway surface fluid and alveolar macrophages, inhibit retroviral gene transfer to airway epithelia. Human alveolar macrophages obtained by bronchoalveolar lavage from normal subjects were left untreated or activated with lipopolysaccharide (LPS) for 3 hr in the presence of subconfluent human bronchial epithelial cells (HBE); than 4 x 10(5) cfu DA-luciferase retrovirus was added. Three days after infection, luciferase activity was measured in cell lysates. When the epithelial cells were co-cultured with LPS-activated macrophages, retroviral gene transfer to HBE cells was reduced by approximately 60%. Nonactivated macrophages decreased the transfection to approximately 55% of control values. In control experiments with either activated or inactivated macrophages but without epithelia, no luciferase activity was detected, suggesting that terminally differentiated alveolar macrophages are not infected by the recombinant retrovirus. Pretreatment of alveolar macrophages with dexamethasone restored gene transfer to approximately 60% of control values. In contrast, incubation of retrovirus with airway surface fluid had no inhibitory effect on gene transfer. These experiments document that AM inhibit retrovirus-mediated gene transfer to airway epithelia in vitro, and may represent a barrier to retroviral gene transfer in vivo. These barriers may be overcome, at least partially, with pharmacological agents.


Assuntos
Técnicas de Transferência de Genes , Pulmão/fisiologia , Macrófagos Alveolares/fisiologia , Macrófagos Alveolares/virologia , Retroviridae/genética , Antineoplásicos Hormonais/farmacologia , Líquidos Corporais , Células Cultivadas , Dexametasona/farmacologia , Relação Dose-Resposta a Droga , Células Epiteliais , Epitélio/fisiologia , Epitélio/virologia , Vetores Genéticos/genética , Vetores Genéticos/farmacologia , Humanos , Pulmão/virologia , Macrófagos Alveolares/efeitos dos fármacos
17.
Transplantation ; 62(12): 1866-71, 1996 Dec 27.
Artigo em Inglês | MEDLINE | ID: mdl-8990378

RESUMO

Gene transfer techniques can be used as a drug delivery system to achieve local immunosuppression. We performed a series of experiments to identify the cationic lipid that most efficiently transfects isolated, cultured, rat hepatocytes; to optimize conditions for efficient transfection; to determine the duration of gene expression in vitro; and finally, to determine the survival of allogeneic hepatocytes transplanted into Nagase rats. Our results suggest that DOTAP is the best cationic lipid for transfection of cultured rat hepatocytes. In addition, the following conditions appear to optimize transfection efficiency: a DNA:DOTAP ratio of 1:6; a 24 exposure time of the hepatocytes to the DNA-DOTAP complex; a DNA dose of 4 microg/35 mm culture plate seeded with 2.5x10(5) rat hepatocytes. When transfected as described above, cultured hepatocytes expressed the hIL-10 gene for approximately 14 days. Accordingly, Nagase rats transplanted with 4x10(7) DOTAP-hIL-10 transfected, allogeneic hepatocytes had an abrupt rise in serum albumin levels that peaked within 7 days of the transplant, decreased abruptly after 15 days, and approached baseline by day 40. In contrast, control animals had a smaller albumin peak that returned to baseline within 10 days (P<0.01). In all animals, serum hIL-10 levels were undetectable when tested. We conclude that DOTAP is the best cationic lipid for transfection of cultured rat hepatocytes. Furthermore, hIL-10 transfected hepatocytes have a prolonged survival in an allogeneic host which is probably limited by loss of gene expression. Further studies using other vectors capable of prolonged gene expression will help determine if indefinite hIL-10 gene expression leads to indefinite graft survival.


Assuntos
Ácidos Graxos Monoinsaturados/farmacologia , Interleucina-10/genética , Transplante de Fígado/imunologia , Fígado/citologia , Compostos de Amônio Quaternário/farmacologia , Animais , Células Cultivadas , DNA/análise , Expressão Gênica , Técnicas de Transferência de Genes , Sobrevivência de Enxerto/efeitos dos fármacos , Humanos , Tolerância Imunológica/efeitos dos fármacos , Lipídeos/genética , Ratos , Ratos Endogâmicos BN , Transfecção/efeitos dos fármacos
18.
Intensive Care Med ; 15(3): 155-9, 1989.
Artigo em Inglês | MEDLINE | ID: mdl-2738219

RESUMO

We have studied the effect of changing ventilatory frequency (VF) of high frequency oscillatory ventilation (HFOV) on fluid filtration rate (FFR) in twelve isolated rabbit lungs perfused at constant blood flow. Mean pulmonary artery pressure (Ppa), mean left atrial pressure (Pla), airway pressure (Paw), pulmonary vascular resistance (PVR), pH, O2 and CO2 partial arterial pressures (PaO2 and PaCO2) and plasma colloid osmotic pressure (COP), were measured. We ventilated the lungs with a modified Bird Mark 7 ventilator which could achieve HFOV (range 5-30 Hertz). In each experiment VF was randomly varied on ten different occasions, maintaining each variation for ten minutes. The first group of six rabbits was ventilated under normal haemodynamic conditions. The other six rabbits were ventilated after the production of hydrostatic lung oedema. Blood gas exchange in both groups of rabbits was satisfactory. There was no statistically significant correlation between VF and FFR. We conclude that variations in VF using HFOV does not alter lung fluid balance in normal and in hydrostatic oedema rabbit lungs.


Assuntos
Líquidos Corporais/fisiologia , Ventilação de Alta Frequência , Pulmão/fisiopatologia , Animais , Filtração , Edema Pulmonar/fisiopatologia , Troca Gasosa Pulmonar , Pressão Propulsora Pulmonar , Coelhos
19.
Intensive Care Med ; 16(2): 89-94, 1990.
Artigo em Inglês | MEDLINE | ID: mdl-2185291

RESUMO

The influence of positive end expiratory pressure (PEEP) on the fluid filtration rate (FFR) in the pulmonary circulation has been the subject of considerable investigation but data are conflicting. We studied twenty-nine isolated rabbit lung preparations, FFR was sensed by a force transducer. Autologous blood was used to prime the perfusion circuit. Hydrostatic oedema was achieved by increasing the left atrial pressure to 16 mmHg. In order to bring about increased membrane permeability oleic acid was injected through the pulmonary artery. Increasing and decreasing levels of PEEP at 0, 5, 10 and 15 cm H2O were each used for ten minutes in each of three experimental models. The FFR, pH, mean pulmonary arterial pressure (MPAP), mean left atrial pressure (MLAP), PaO2, PaCO2 and oncotic pressure were measured in each experiment. There was a significant correlation between PEEP and FFR (+0.94) in non-oedema lungs. With no PEEP the FFR was 0 g/min and with 15 cm of PEEP it increased to 0.07 g/min, on removing the PEEP the FFR returned to 0 g/min. In the hydrostatic lung oedema model the correlation was also significant but negative (r = -0.94). With no PEEP the FFR was 0.33 g/min, with PEEP of 15 cm H2O it decreased to 0.08 g/min. No correlation between PEEP and FFR was found in the oleic acid preparation. In the normal lung PEEP increases capillary hydrostatic pressure and total lung vascular area and decreases interstitial pressure. It is by these mechanisms that PEEP causes an increase in FFR. In the hydrostatic oedema model PEEP decreases FFR by increasing the interstitial pressure and by decreasing the total lung vascular area. In the oleic acid preparation the coefficient of filtration is so large that small changes in pressure or vascular area do not modify the FFR. We suggest that PEEP may be beneficial by decreasing FFR in hydrostatic lung oedema, but it may increase the FFR in the normal lung, while having no effect in oleic acid lung injury.


Assuntos
Respiração com Pressão Positiva , Circulação Pulmonar , Edema Pulmonar/fisiopatologia , Resistência das Vias Respiratórias , Animais , Permeabilidade da Membrana Celular , Modelos Animais de Doenças , Ácidos Oleicos/efeitos adversos , Edema Pulmonar/induzido quimicamente , Edema Pulmonar/terapia , Pressão Propulsora Pulmonar , Coelhos
20.
Laryngoscope ; 111(7): 1290-6, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11568557

RESUMO

OBJECTIVES/HYPOTHESIS: Adenoviral-mediated gene transfer offers a potential new treatment strategy for squamous cell cancer of the head and neck (SCCHN). Initial studies on some SCCHN cell lines have shown that these cells can be resistant to adenovirus-mediated gene transfer, requiring large amounts of vector and long infection times. The objectives of this study were to identify the barriers to gene transfer in three SCCHN lines, FaDu, SCC-9, and SCC-15, and to develop a method to circumvent the obstacles. We hypothesized that a low expression of adenovirus receptors may limit adenovirus infection and this may be overcome by using adenovirus complexed with calcium phosphate coprecipitates. METHODS: Using standard cell and molecular biology techniques, the infectability of SCCHN cells was investigated. RESULTS: Using Cy3-labeled adenovirus, we found minimal binding of adenovirus to FaDu cells and variable levels of binding among SCC-9 and SCC-15 cells. Northern blot analysis indicated that messenger RNA (mRNA) transcripts for coxsackie-adenovirus receptor, which binds adenovirus, were absent in FaDu cells but present in SCC-9 and SCC-15 cells. Integrin alphavbeta5, which binds and facilitates internalization of adenovirus, were expressed at low levels in all three cell types. We overcame these barriers by using adenovirus complexed with calcium phosphate precipitates. Total transgene expression and the number of cells expressing transgene were increased in all three cancer lines using adenovirus complexed with calcium phosphate precipitates compared with adenovirus that was not complexed. CONCLUSIONS: Data in the present study suggest that adenovirus-mediated gene transfer to SCCHN cell lines is a result of limited viral receptors. Delivering adenovirus in a calcium phosphate coprecipitate enhanced gene transfer and, perhaps, the therapeutic index.


Assuntos
Adenoviridae/genética , Fosfatos de Cálcio/farmacologia , Carcinoma de Células Escamosas/terapia , Técnicas de Transferência de Genes , Vetores Genéticos , Neoplasias de Cabeça e Pescoço/terapia , Animais , Northern Blotting , Linhagem Celular , Enterovirus , Células HeLa , Humanos , Imuno-Histoquímica , Integrinas/genética , Camundongos , Microscopia Confocal , RNA Mensageiro/genética , Receptores Virais/genética , Fatores de Tempo , Transgenes , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA