Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Am J Transplant ; 13(8): 2137-45, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23710593

RESUMO

We conducted a retrospective study of deep surgical site infections (SSIs) among consecutive patients who underwent lung transplantation (LTx) at a single center from 2006 through 2010. Thirty-one patients (5%) developed SSIs at median 25 days after LTx. Empyema was most common (42%), followed by surgical wound infections (29%), mediastinitis (16%), sternal osteomyelitis (6%), and pericarditis (6%). Pathogens included Gram-positive bacteria (41%), Gram-negative bacteria (41%), fungi (10%) and Mycobacterium abscessus, Mycoplasma hominis and Lactobacillus sp. (one each). Twenty-three percent of SSIs were due to pathogens colonizing recipients' native lungs at time of LTx, suggesting surgical seeding as a source. Patient-related independent risk factors for SSIs were diabetes and prior cardiothoracic surgery; procedure-related independent risk factors were LTx from a female donor, prolonged ischemic time and number of perioperative red blood cell transfusions. Mediastinitis and sternal infections were not observed among patients undergoing minimally invasive LTx. SSIs were associated with 35% mortality at 1 year post-LTx. Lengths of stay and mortality in-hospital and at 6 months and 1 year were significantly greater for patients with SSIs other than empyema. In conclusion, deep SSIs were uncommon, but important complications in LTx recipients because of their diverse microbiology and association with increased mortality.


Assuntos
Rejeição de Enxerto/mortalidade , Infecções por Bactérias Gram-Negativas/epidemiologia , Infecções por Bactérias Gram-Positivas/epidemiologia , Pneumopatias/complicações , Transplante de Pulmão/efeitos adversos , Complicações Pós-Operatórias , Infecção da Ferida Cirúrgica/epidemiologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Seguimentos , Rejeição de Enxerto/etiologia , Bactérias Gram-Negativas/patogenicidade , Infecções por Bactérias Gram-Negativas/microbiologia , Infecções por Bactérias Gram-Negativas/mortalidade , Bactérias Gram-Positivas/patogenicidade , Infecções por Bactérias Gram-Positivas/microbiologia , Infecções por Bactérias Gram-Positivas/mortalidade , Humanos , Pneumopatias/mortalidade , Pneumopatias/cirurgia , Masculino , Pessoa de Meia-Idade , Prognóstico , Estudos Retrospectivos , Fatores de Risco , Infecção da Ferida Cirúrgica/etiologia , Infecção da Ferida Cirúrgica/mortalidade , Taxa de Sobrevida , Estados Unidos/epidemiologia , Adulto Jovem
2.
Eur Respir J ; 35(4): 781-6, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19717485

RESUMO

Biomarkers providing in vivo quantification of the basic elements of cystic fibrosis (CF) lung disease are needed. A study was performed to determine whether the absorption of a small radiolabelled hydrophilic molecule (Indium-111 (In-)DTPA) would be increased in CF airways. DTPA clearance has been used previously to assess epithelial permeability and may also be useful for quantifying liquid absorption. The absorptive clearance rate of DTPA was quantified in 10 CF and 11 control subjects using a novel aerosol technique. Subjects inhaled an aerosol containing nonabsorbable technetium-99m sulfur colloid (Tc-SC) particles and In-DTPA. Tc-SC clearance from the lung is exclusively mucociliary, while In-DTPA is cleared by both absorption and mucociliary clearance. The difference between the In-DTPA and Tc-SC clearance rates estimates In-DTPA absorption. Tc-SC (mucociliary) clearance was similar in central and peripheral zones in CF and non-CF lungs. Total In-DTPA clearance was increased in both zones in CF lungs. The absorptive component of In-DTPA clearance was increased in the airway-dominated central lung zones in CF (42% x h( -1) versus 32% x h(-1), p = 0.03). The absorption of In-DTPA is increased in the CF airway. Further study is needed to understand the relative roles of fluid absorption, inflammation and other mechanisms potentially affecting epithelial permeability and DTPA absorption.


Assuntos
Fibrose Cística/diagnóstico por imagem , Fibrose Cística/metabolismo , Radioisótopos de Índio , Ácido Pentético , Mucosa Respiratória/metabolismo , Adolescente , Adulto , Aerossóis , Biomarcadores/metabolismo , Feminino , Humanos , Radioisótopos de Índio/farmacocinética , Modelos Lineares , Masculino , Ácido Pentético/farmacocinética , Projetos Piloto , Cintilografia , Testes de Função Respiratória , Adulto Jovem
3.
Am J Physiol Lung Cell Mol Physiol ; 281(1): L16-23, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11404240

RESUMO

To test the hypothesis that Na+ transport in human bronchial epithelial (HBE) cells is regulated by a protease-mediated mechanism, we investigated the effects of BAY 39-9437, a recombinant Kunitz-type serine protease inhibitor, on amiloride-sensitive short-circuit current of normal [non-cystic fibrosis (CF) cells] and CF HBE cells. Mucosal treatment of non-CF and CF HBE cells with BAY 39-9437 decreased the short-circuit current, with a half-life of approximately 45 min. At 90 min, BAY 39-9437 (470 nM) reduced Na+ transport by approximately 70%. The inhibitory effect of BAY 39-9437 was concentration dependent, with a half-maximal inhibitory concentration of approximately 25 nM. Na+ transport was restored to control levels, with a half-life of approximately 15 min, on washout of BAY 39-9437. In addition, trypsin (1 microM) rapidly reversed the inhibitory effect of BAY 39-9437. These data indicate that Na+ transport in HBE cells is activated by a BAY 39-9437-inhibitable, endogenously expressed serine protease. BAY 39-9437 inhibition of this serine protease maybe of therapeutic potential for the treatment of Na+ hyperabsorption in CF.


Assuntos
Brônquios/metabolismo , Fibrose Cística/metabolismo , Inibidores de Proteases/farmacologia , Proteínas Recombinantes/farmacologia , Sódio/metabolismo , Transporte Biológico/efeitos dos fármacos , Células Cultivadas , Células Epiteliais/metabolismo , Humanos , Valores de Referência
4.
Am J Respir Cell Mol Biol ; 24(5): 556-62, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11350824

RESUMO

Significant progress has been made toward identifying growth factors that display autocrine or paracrine effects on the growth of lung cancer cells. Determining the in vivo relevance of specific growth factors on lung tumor formation, however, has not often been demonstrated in laboratory models. Although hepatocyte growth factor (HGF) has been shown to have mitogenic and motogenic effects on human lung cancer cells in vitro, and to have prognostic importance in patients with lung cancer, the effects of HGF on tumor behavior in vivo remain unknown. We therefore developed an airway tumor xenograft model that allowed us to test the hypothesis that HGF promotes human non-small cell lung cancer (NSCLC) growth in vivo. Human airway tumor xenografts were created in Severe Combined Immunodeficient mice by injecting human lung adenocarcinoma cells into human bronchial segments. After determining the optimal times for tumor-cell injection and the time course of tumor growth, we evaluated the effects of HGF on tumor growth by injecting recombinant HGF, or saline as a control, into the lumen of tumor xenografts for 10 consecutive days. Histologic evaluation 2 to 3 wk later revealed that the HGF-injected xenografts had a significantly greater tumor volume and more tumor cells were located in the submucosal space than were found in the saline-injected xenografts. These data demonstrate the usefulness of this novel in vivo model to study NSCLC, and show that HGF promotes both the growth and invasion of human lung cancer in vivo.


Assuntos
Adenocarcinoma/metabolismo , Fator de Crescimento de Hepatócito/metabolismo , Neoplasias Pulmonares/metabolismo , Adenocarcinoma/patologia , Animais , Divisão Celular/efeitos dos fármacos , Modelos Animais de Doenças , Fator de Crescimento de Hepatócito/administração & dosagem , Humanos , Imuno-Histoquímica , Instilação de Medicamentos , Neoplasias Pulmonares/patologia , Camundongos , Camundongos SCID , Transplante de Neoplasias , Mucosa Respiratória/patologia , Sensibilidade e Especificidade , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
5.
Am J Physiol Cell Physiol ; 279(2): C461-79, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10913013

RESUMO

Forskolin, UTP, 1-ethyl-2-benzimidazolinone (1-EBIO), NS004, 8-methoxypsoralen (Methoxsalen; 8-MOP), and genistein were evaluated for their effects on ion transport across primary cultures of human bronchial epithelium (HBE) expressing wild-type (wt HBE) and DeltaF508 (DeltaF-HBE) cystic fibrosis transmembrane conductance regulator. In wt HBE, the baseline short-circuit current (I(sc)) averaged 27.0 +/- 0.6 microA/cm(2) (n = 350). Amiloride reduced this I(sc) by 13.5 +/- 0.5 microA/cm(2) (n = 317). In DeltaF-HBE, baseline I(sc) was 33.8 +/- 1.2 microA/cm(2) (n = 200), and amiloride reduced this by 29.6 +/- 1.5 microA/cm(2) (n = 116), demonstrating the characteristic hyperabsorption of Na(+) associated with cystic fibrosis (CF). In wt HBE, subsequent to amiloride, forskolin induced a sustained, bumetanide-sensitive I(sc) (DeltaI(sc) = 8.4 +/- 0.8 microA/cm(2); n = 119). Addition of acetazolamide, 5-(N-ethyl-N-isopropyl)-amiloride, and serosal 4, 4'-dinitrostilben-2,2'-disulfonic acid further reduced I(sc), suggesting forskolin also stimulates HCO(3)(-) secretion. This was confirmed by ion substitution studies. The forskolin-induced I(sc) was inhibited by 293B, Ba(2+), clofilium, and quinine, whereas charybdotoxin was without effect. In DeltaF-HBE the forskolin I(sc) response was reduced to 1.2 +/- 0.3 microA/cm(2) (n = 30). In wt HBE, mucosal UTP induced a transient increase in I(sc) (Delta I(sc) = 15. 5 +/- 1.1 microA/cm(2); n = 44) followed by a sustained plateau, whereas in DeltaF-HBE the increase in I(sc) was reduced to 5.8 +/- 0. 7 microA/cm(2) (n = 13). In wt HBE, 1-EBIO, NS004, 8-MOP, and genistein increased I(sc) by 11.6 +/- 0.9 (n = 20), 10.8 +/- 1.7 (n = 18), 10.0 +/- 1.6 (n = 5), and 7.9 +/- 0.8 microA/cm(2) (n = 17), respectively. In DeltaF-HBE, 1-EBIO, NS004, and 8-MOP failed to stimulate Cl(-) secretion. However, addition of NS004 subsequent to forskolin induced a sustained Cl(-) secretory response (2.1 +/- 0.3 microA/cm(2), n = 21). In DeltaF-HBE, genistein alone stimulated Cl(-) secretion (2.5 +/- 0.5 microA/cm(2), n = 11). After incubation of DeltaF-HBE at 26 degrees C for 24 h, the responses to 1-EBIO, NS004, and genistein were all potentiated. 1-EBIO and genistein increased Na(+) absorption across DeltaF-HBE, whereas NS004 and 8-MOP had no effect. Finally, Ca(2+)-, but not cAMP-mediated agonists, stimulated K(+) secretion across both wt HBE and DeltaF-HBE in a glibenclamide-dependent fashion. Our results demonstrate that pharmacological agents directed at both basolateral K(+) and apical Cl(-) conductances directly modulate Cl(-) secretion across HBE, indicating they may be useful in ameliorating the ion transport defect associated with CF.


Assuntos
Brônquios/metabolismo , Canais de Cloreto/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Células Epiteliais/metabolismo , Canais de Potássio/metabolismo , Benzimidazóis/farmacologia , Brônquios/efeitos dos fármacos , Células Cultivadas , Canais de Cloreto/efeitos dos fármacos , Fibrose Cística/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/agonistas , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Fármacos Dermatológicos/farmacologia , Inibidores Enzimáticos/farmacologia , Células Epiteliais/efeitos dos fármacos , Furocumarinas/farmacologia , Genisteína/farmacologia , Humanos , Transporte de Íons/efeitos dos fármacos , Transporte de Íons/fisiologia , Canais de Potássio/efeitos dos fármacos
6.
J Pharmacol Exp Ther ; 292(2): 778-87, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10640318

RESUMO

We previously demonstrated that 1-ethyl-2-benzimidazolone (1-EBIO) directly activates basolateral membrane calcium-activated K(+) channels (K(Ca)), thereby stimulating Cl(-) secretion across several epithelia. In our pursuit to identify potent modulators of Cl(-) secretion that may be useful to overcome the Cl(-) secretory defect in cystic fibrosis (CF), we have identified chlorzoxazone [5-chloro-2(3H)-benzoxazolone], a clinically used centrally acting muscle relaxant, as a stimulator of Cl(-) secretion in several epithelial cell types, including T84, Calu-3, and human bronchial epithelium. The Cl(-) secretory response induced by chlorzoxazone was blocked by charybdotoxin (CTX), a known blocker of K(Ca). In nystatin-permeabilized monolayers, chlorzoxazone stimulated a basolateral membrane I(K), which was inhibited by CTX and also stimulated an apical I(Cl) that was inhibited by glibenclamide, indicating that the G(Cl) responsible for this I(Cl) may be cystic fibrosis transmembrane conductance regulator (CFTR). In membrane vesicles prepared from T84 cells, chlorzoxazone stimulated (86)Rb(+) uptake in a CTX-sensitive manner. In excised, inside-out patches, chlorzoxazone activated an inwardly-rectifying K(+) channel, which was inhibited by CTX. 6-Hydroxychlorzoxazone, the major metabolite of chlorzoxazone, did not activate K(Ca), whereas zoxazolamine (2-amino-5-chlorzoxazole) showed a similar response profile as chlorzoxazone. In normal human nasal epithelium, chlorzoxazone elicited hyperpolarization of the potential difference that was similar in magnitude to isoproterenol. However, in the nasal epithelium of CF patients with the DeltaF508 mutation of CFTR, there was no detectable Cl(-) secretory response to chlorzoxazone. These studies demonstrate that chlorzoxazone stimulates transepithelial Cl(-) secretion in normal airway epithelium in vitro and in vivo, and suggest that stimulation requires functional CFTR in the epithelia.


Assuntos
Ânions/metabolismo , Brônquios/metabolismo , Cloro/metabolismo , Clorzoxazona/farmacologia , Mucosa Nasal/efeitos dos fármacos , Amilorida/farmacologia , Bumetanida/farmacologia , Membrana Celular/efeitos dos fármacos , Células Cultivadas , Charibdotoxina/farmacologia , Clorzoxazona/análogos & derivados , Colforsina/farmacologia , Fibrose Cística/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Interações Medicamentosas , Epitélio/metabolismo , Glibureto/farmacologia , Humanos , Isoproterenol/farmacologia , Nistatina/farmacologia , Bloqueadores dos Canais de Potássio , Rubídio/farmacocinética , Zoxazolamina/farmacologia
7.
Am J Physiol ; 276(4): C827-37, 1999 04.
Artigo em Inglês | MEDLINE | ID: mdl-10199813

RESUMO

Ca2+-mediated agonists, including UTP, are being developed for therapeutic use in cystic fibrosis (CF) based on their ability to modulate alternative Cl- conductances. As CF is also characterized by hyperabsorption of Na+, we determined the effect of mucosal UTP on transepithelial Na+ transport in primary cultures of human bronchial epithelia (HBE). In symmetrical NaCl, UTP induced an initial increase in short-circuit current (Isc) followed by a sustained inhibition. To differentiate between effects on Na+ absorption and Cl- secretion, Isc was measured in the absence of mucosal and serosal Cl- (INa). Again, mucosal UTP induced an initial increase and then a sustained decrease that reduced amiloride-sensitive INa by 73%. The Ca2+-dependent agonists histamine, bradykinin, serosal UTP, and thapsigargin similarly induced sustained inhibition (62-84%) of INa. Mucosal UTP induced similar sustained inhibition (half-maximal inhibitory concentration 296 nM) of INa in primary cultures of human CF airway homozygous for the DeltaF508 mutation. BAPTA-AM blunted UTP-dependent inhibition of INa, but inhibitors of protein kinase C (PKC) and phospholipase A2 had no effect. Indeed, direct activation of PKC by phorbol 12-myristate 13-acetate failed to inhibit Na+ absorption. Apyrase, a tri- and diphosphatase, did not reverse inhibitory effects of UTP on INa, suggesting a long-term inhibitory effect of UTP that is independent of receptor occupancy. After establishment of a mucosa-to-serosa K+ concentration gradient and permeabilization of the mucosal membrane with nystatin, mucosal UTP induced an initial increase in K+ current followed by a sustained inhibition. We conclude that increasing cellular Ca2+ induces a long-term inhibition of transepithelial Na+ transport across normal and CF HBE at least partly due to downregulation of a basolateral membrane K+ conductance. Thus UTP may have a dual therapeutic effect in CF airway: 1) stimulation of a Cl- secretory response and 2) inhibition of Na+ transport.


Assuntos
Brônquios/fisiopatologia , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Fibrose Cística/fisiopatologia , Sódio/metabolismo , Uridina Trifosfato/farmacologia , Amilorida/farmacologia , Transporte Biológico/efeitos dos fármacos , Bumetanida , Cálcio/fisiologia , Células Cultivadas , Cloretos/metabolismo , Fibrose Cística/genética , Regulador de Condutância Transmembrana em Fibrose Cística/fisiologia , Histamina/farmacologia , Homozigoto , Humanos , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Mucosa/efeitos dos fármacos , Mucosa/fisiopatologia , Deleção de Sequência , Acetato de Tetradecanoilforbol/farmacologia
8.
Physiol Rev ; 79(1 Suppl): S215-55, 1999 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9922383

RESUMO

Role of CFTR in Airway Disease. Physiol. Rev. 79, Suppl.: S215-S255, 1999. - Cystic fibrosis (CF) is caused by mutations in the gene encoding the CF transmembrane conductance regulator (CFTR), which accounts for the cAMP-regulated chloride conductance of airway epithelial cells. Lung disease is the chief cause of morbidity and mortality in CF patients. This review focuses on mechanisms whereby the deletion or impairment of CFTR chloride channel function produces lung disease. It examines the major themes of the channel hypothesis of CF, which involve impaired regulation of airway surface fluid volume or composition. Available evidence indicates that the effect of CFTR deletion alters physiological functions of both surface and submucosal gland epithelia. At the airway surface, deletion of CFTR causes hyperabsorption of sodium chloride and a reduction in the periciliary salt and water content, which impairs mucociliary clearance. In submucosal glands, loss of CFTR-mediated salt and water secretion compromises the clearance of mucins and a variety of defense substances onto the airway surface. Impaired mucociliary clearance, together with CFTR-related changes in the airway surface microenvironment, leads to a progressive cycle of infection, inflammation, and declining lung function. Here, we provide the details of this pathophysiological cascade in the hope that its understanding will promote the development of new therapies for CF.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Fibrose Cística/fisiopatologia , Pulmão/fisiopatologia , Animais , Líquidos Corporais/metabolismo , Fibrose Cística/genética , Fibrose Cística/metabolismo , Fibrose Cística/patologia , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Humanos , Inflamação , Pulmão/metabolismo , Depuração Mucociliar/fisiologia , Infecções Respiratórias
9.
Am J Physiol ; 273(5): L913-20, 1997 11.
Artigo em Inglês | MEDLINE | ID: mdl-9374717

RESUMO

Altered glycosylation of mucus and membrane glycoconjugates could explain reported differences in binding of bacterial pathogens to cystic fibrosis (CF) versus normal tissue. However, because bacteria can alter cell surface glycoconjugates, it is not possible to assess the role of cystic fibrosis transmembrane conductance regulators (CFTR) in glycosylation in these studies. To address this issue, we have developed quantitative lectin binding assays to compare cell surface glycosylation in well-matched immortalized CF cells and rescued cell lines. The CF airway bronchial epithelial cell line IB3-1 consistently bound more peanut agglutinin (PNA) than its clonal derivative S9, which stably expresses functional wild-type CFTR. Pretreatment with neuraminidase increased PNA binding and abolished the difference between the two cell lines. However, infection of the IB3-1 cells with a replication-deficient recombinant adenovirus encoding CFTR restored CFTR function but did not alter PNA binding to cells. In contrast, treatment with the weak base ammonium chloride increased PNA binding to both cell lines as expected. Our data show that even clonally related CF and rescued cells can exhibit significant differences in carbohydrate processing. Although the differences that we found are consistent with the proposed role for CFTR in modulating intraorganellar pH, our data strongly suggest that they are CFTR independent. These studies add a cautionary note to the interpretation of differences in glycosylation between CF and normal primary tissues and immortalized cells.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Fibrose Cística/metabolismo , Adenoviridae , Animais , Sítios de Ligação , Brônquios , Linhagem Celular , Permeabilidade da Membrana Celular , AMP Cíclico/metabolismo , Células Epiteliais , Vetores Genéticos , Glicosilação , Humanos , Neuraminidase , Aglutinina de Amendoim , Proteínas Recombinantes/metabolismo , Spodoptera , Transfecção , Conjugado Aglutinina do Germe de Trigo-Peroxidase do Rábano Silvestre
10.
Am J Respir Cell Mol Biol ; 17(4): 422-35, 1997 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9376117

RESUMO

Recombinant adenoviruses are currently being evaluated as gene transfer vectors for the treatment of airway diseases. Recent evidence indicates that gene transfer to differentiated airway epithelial cells is inefficient. We hypothesized that apical membrane glycoconjugates, such as the transmembrane mucin MUC1, reduce the efficiency of adenovirus-mediated gene transfer. To address this, studies were performed in primary bronchial epithelial and Madin Darby canine kidney (MDCK) cells transduced to express human MUC1. Colocalization of MUC1 and an adenoviral lacZ transgene in the bronchial epithelial cells revealed that at several multiplicities of infection, the percentage of cells expressing lacZ was five-fold less in MUC1-expressing cells. Moreover, lacZ expression was three- to eight-fold lower in MUC1-expressing than in control MDCK cells, demonstrating that MUC1 interferes with gene transfer and is not merely a phenotypic marker of a cell that is refractory to adenovirus infection. Neuraminidase pretreatment of cells to remove sialic acid residues prior to viral adsorption increased the efficiency of gene transfer two- to five-fold in human airway and MDCK cells, and in a xenograft model of human airway. This effect was also observed in cultured cells that do not express MUC1, suggesting that other sialylated glycoconjugates impact on the efficiency of gene transfer. An inhibitory effect of negatively charged glycoconjugates on adenovirus binding was further supported by the finding that adsorption of adenovirus with a polycation significantly increased gene transfer efficiency. These data demonstrate for the first time that sialoglycoconjugates on epithelial cells reduce the efficiency of adenovirus-mediated gene transfer.


Assuntos
Brônquios/metabolismo , Regulação da Expressão Gênica , Técnicas de Transferência de Genes , Mucina-1/genética , Adenoviridae , Animais , Células Cultivadas , Cães , Epitélio/metabolismo , Vetores Genéticos , Glicoconjugados/genética , Glicoconjugados/metabolismo , Humanos , Mucina-1/metabolismo
11.
Am J Respir Crit Care Med ; 156(2 Pt 1): 358-66, 1997 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9279210

RESUMO

Bombesin-like peptides (BLPs) are important regulators of lung development and may also act as autocrine growth factors in lung tumors. We have previously demonstrated expression of mRNA for the three BLP receptor subtypes (neuromedin B [NMB]) receptor, gastrin-releasing peptide [GRP] receptor, and bombesin receptor subtype 3 [BRS-3]) in human non-small cell lung carcinoma (NSCLC) cell lines and bronchial biopsies using the reverse transcription-polymerase chain reaction (RT-PCR; DeMichele, et al. Am. J. Respir. Cell Mol. Biol. 1994; 11:66-74). We have also previously found that growth responses to BLPs could be elicited in some, but not all, cultures of human bronchial epithelial (HBE) cells (Siegfried, et al. Anat. Rec. 1993; 236:241-247). In this report, we utilized RT-PCR to demonstrate mRNA expression of BLP receptor subtypes in cultured HBE cells and also assessed the response of these cultures to BLPs in proliferation assays. The pattern of mRNA expression was correlated with proliferative response, and the results were also analyzed in relation to smoking history and pulmonary function of the subjects studied. Our results suggest that expression of mRNA for the GRP receptor is associated with a long smoking history (> 25 pack-years [PY], p = 0.02). This association was related to past tobacco exposure, regardless of whether the subjects were still active smokers at the time of tissue procurement. Responsiveness to GRP and NMB in proliferation assays was also found only in those HBE cultures with expression of mRNA for at least one of the known receptors for BLPs, and there was a significant association between expression of mRNA for the GRP receptor and proliferative response to both GRP and NMB (p = 0.048). HBE cultures from subjects with a greater than 25 PY smoking history were also more likely to respond to BLPs in the proliferation assays than cells from subjects with less than a 25 PY history (10 of 16 versus 1 of 7, p = 0.06). Cultures of HBE cells from four of the five subjects with severe obstructive lung disease gave a positive response to GRP and NMB in proliferation assays, compared to five of fifteen without severe obstructive lung disease, but this difference was not significant (p = 0.13). These results suggest there is an increased likelihood of expression of the GRP receptor mRNA in the respiratory epithelium of some individuals with a history of prolonged tobacco exposure, and that expression of the GRP receptor mRNA is accompanied by responsiveness to the mitogenic effects of BLPs. These effects appear to persist after smoking cessation.


Assuntos
Bombesina/farmacologia , Brônquios/efeitos dos fármacos , Nicotiana , Plantas Tóxicas , RNA Mensageiro/efeitos dos fármacos , Receptores da Bombesina/efeitos dos fármacos , Fumaça/efeitos adversos , Adulto , Idoso , Bombesina/efeitos dos fármacos , Bombesina/metabolismo , Brônquios/citologia , Brônquios/metabolismo , Divisão Celular/efeitos dos fármacos , Células Cultivadas , Ensaio de Unidades Formadoras de Colônias , Células Epiteliais , Epitélio/efeitos dos fármacos , Epitélio/metabolismo , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase/métodos , RNA Mensageiro/metabolismo , Receptores da Bombesina/metabolismo
12.
Am J Physiol ; 273(1 Pt 1): L256-63, 1997 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-9252563

RESUMO

Airway epithelium is subject to injury during inflammation and exposure to a variety of inhaled and infectious agents. Little is known about the expression of integrins during human airway epithelial regeneration and differentiation after injury. We therefore characterized integrin subunit expression after mechanical injury in an in vivo xenograft model of human bronchial epithelium. On the migrating cells at the edges of surface epithelial wounds, there was increased expression of the alpha v-, beta 5-, beta 6-, and alpha 5-integrin subunits. During the later phase of repair, the increased expression of alpha v-, beta 5-, and beta 6-subunits persisted, but the expression of the beta 8-subunits was restricted to basal cells. In addition, there was a redistribution of the alpha 2- and alpha 6-collagen/laminin-binding integrins to suprabasal epithelial layers. There was no expression of the beta 3- or alpha 4-integrin subunit on reparative epithelium. A similar upregulation of alpha v-, beta 5-, and beta 6-integrin receptor subunits was observed in areas of undifferentiated airway from cystic fibrosis patients. Injured epithelium was found to be significantly more susceptible to gene transfer with a recombinant adenovirus, suggesting that the increased integrin expression has implications for the acquisition of adenovirus infections and for lung-directed gene therapy.


Assuntos
Brônquios/fisiologia , Cadeias beta de Integrinas , Integrinas/biossíntese , Transplante Heterólogo/fisiologia , Cicatrização/imunologia , Animais , Antígenos CD/biossíntese , Brônquios/lesões , Brônquios/transplante , Fibrose Cística/imunologia , Fibrose Cística/patologia , Epitélio/lesões , Epitélio/fisiologia , Epitélio/transplante , Humanos , Integrina alfa5 , Integrina alfaV , Camundongos , Camundongos SCID , Transplante Heterotópico , Regulação para Cima
14.
Gene Ther ; 4(1): 32-8, 1997 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9068793

RESUMO

Recombinant adenoviruses are being developed for gene therapy for cystic fibrosis and other lung diseases, and for prevention and treatment of vascular thrombosis. A major limitation to the clinical utility of adenoviruses is the low efficiency of gene transfer achieved in vivo. In addition, little is known about the initial interactions between adenoviruses and the target cell. To address the hypothesis that the negative charge presented by membrane glycoproteins reduces the efficiency of adenovirus-mediated gene transfer, primary cultures of human airway, Madin-Darby canine kidney cells, an immortalized cystic fibrosis airway epithelial cell line, and primary cultures of sheep pulmonary artery endothelium were infected with recombinant adenovirus containing the E. coli lacZ reporter gene (Ad2 beta gal2) in the presence of various polyions. For each cell type, adsorption of Ad2 beta gal2 in the presence of the polycations polybrene, protamine, DEAE-dextran, and poly-L-lysine significantly increased the percentage of cells that express lacZ. The polyanion heparin did not significantly alter gene transfer efficiency, but completely abrogated the effects of polycations. These data provide evidence that negatively charged moieties on the cell surface reduce the efficiency of adenovirus-mediated gene transfer, and that alteration of the charge interaction between adenoviruses and the cell surface may improve the potential clinical application of these vectors.


Assuntos
Adenoviridae , Cátions , Técnicas de Transferência de Genes , Vetores Genéticos , Animais , Células Cultivadas , Fibrose Cística/terapia , Cães , Endotélio Vascular , Epitélio , Escherichia coli , Expressão Gênica , Genes Reporter , Humanos , Rim , Óperon Lac , Ovinos , Trombose/terapia , Transgenes
15.
Am J Respir Cell Mol Biol ; 14(1): 1-18, 1996 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-8534480

RESUMO

Gene therapy is the treatment of any disorder or pathophysiologic state based upon the transfer of genetic information. The lung represents a major target of gene therapy for the treatment of genetic disorders such as cystic fibrosis and alpha 1-antitrypsin deficiency. Other diseases are also being targeted, including pulmonary inflammation, surfactant deficiency, pulmonary hypertension, lung cancer, and malignant mesothelioma. This review will examine some general concepts regarding gene transfer and gene therapy, provide an overview of the current vectors being developed to achieve safe and efficient gene transfer, and summarize the ongoing work to apply this new technology to the treatment of both inherited and acquired pulmonary diseases. Although tremendous progress has been made in the ability to successfully transfer genes to cells, there are several unresolved problems limiting the clinical application of this technology to human pulmonary disease. However, as vector technology evolves, gene therapy may become a reality for a number of lung diseases.


Assuntos
Terapia Genética , Pneumopatias/terapia , Animais , Técnicas de Transferência de Genes , Vetores Genéticos , Humanos , Pneumopatias/genética , Vírus/genética
16.
Curr Opin Pulm Med ; 1(6): 435-43, 1995 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9363079

RESUMO

In recent years, several functions of the cystic fibrosis transmembrane conductance regulator have been discovered, yet the pathophysiology of the pulmonary disease in cystic fibrosis remains unclear. At the cellular level, functions of this protein include regulation of chloride and sodium transport at the cell membrane and in intracellular organelles, regulation of protein trafficking, and posttranslational processing of glycoconjugates. Elucidation of these functions has led to several hypotheses to account for how defects in the cystic fibrosis transmembrane conductance regulator produce pulmonary disease, but a clear understanding of the pathophysiologic links between the cellular functions of the cystic fibrosis transmembrane conductance regulator and organ dysfunction has been hampered by the lack of ideal model systems. Current evidence suggests that defects in the cystic fibrosis transmembrane conductance regulator lead to alterations in periciliary fluid homeostasis, mucus hydration, mucin secretion, and apical membrane protein structure. In turn, these alterations impair mucociliary clearance and promote bacterial infection, which then leads to chronic airway inflammation and the development of bronchiectasis.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/genética , Fibrose Cística/genética , Mutação , Fibrose Cística/fisiopatologia , Regulador de Condutância Transmembrana em Fibrose Cística/fisiologia , Humanos , Pneumopatias/genética , Pneumopatias/fisiopatologia
17.
J Clin Immunol ; 15(6 Suppl): 122S-129S, 1995 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-8613485

RESUMO

Endothelial cell-leukocyte interactions involve multiple cell adhesion molecules acting in a programmed and sequential manner to create a leukocyte-endothelial cell adhesion cascade. To understand this process fully, in vivo models are needed. To accomplish this, we have transplanted pieces of normal human tissues onto immunodeficient mice to create chimeric animals. In one model, human skin is grafted and closely resembles normal skin histologically. The grafts retain their human vasculature and show low baseline expression of E-selectin, vascular cell adhesion molecule-1, and intercellular cell adhesion molecule-1. After intradermal injection of human cytokines, these cell adhesion molecules are markedly upregulated and an active inflammatory reaction ensues, with migration of murine leukocytes. Intravenous injection of an anti-human E-selectin antibody completely inhibits leukocyte accumulation induced by tumor necrosis factor-alpha but only partially inhibits leukotriene B4-induced inflammation. In a second model, human bronchus was successfully transplanted heterotopically into severe combined immunodeficient mice. Injection of tumor necrosis factor induced upregulation of E-selectin, intercellular cell adhesion molecule-1, and vascular cell adhesion molecule-1 in the submucosal microvessels, with slightly different kinetics than in the skin. In conclusion, human-severe combined immunodeficient chimeric mice represent a useful model system to study the regulation and function of human cell adhesion molecules in an in vivo setting.


Assuntos
Moléculas de Adesão Celular/fisiologia , Citocinas/fisiologia , Animais , Brônquios/transplante , Movimento Celular/fisiologia , Citocinas/administração & dosagem , Selectina E/fisiologia , Humanos , Leucócitos/fisiologia , Leucotrieno B4/administração & dosagem , Camundongos , Camundongos SCID , Microcirculação , Transplante de Pele , Quimeras de Transplante , Fator de Necrose Tumoral alfa/administração & dosagem
18.
Gene Ther ; 2(5): 344-50, 1995 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-7671110

RESUMO

In utero somatic gene transfer may be a useful therapeutic strategy for a variety of inherited disorders. In the present study, we demonstrate transgene expression in the airways of fetal lamb lungs, 2-3 weeks after injection of Moloney murine leukemia retrovirus based vectors containing cDNA for beta-galactosidase (lacZ) or human interleukin receptor antagonist protein (IRAP), into the fluid filled future airspace of fully catheterized twin fetal lambs (104-117 days gestational age; term 147 days). Expression of lacZ or IRAP was limited to the twin that received the respective vector and was apparent, at light microscopic level, in the epithelium and submucosal space of proximal airways, and to a lesser extent, in the respiratory epithelium of the distal airways. These data demonstrate for the first time that transfer of foreign DNA to fetal lung can be accomplished. These findings support the use of retroviral vectors for somatic lung DNA transfer and suggest that inherited disorders such as cystic fibrosis may be approached therapeutically via gene transfer, in utero.


Assuntos
Técnicas de Transferência de Genes , Pulmão/metabolismo , Retroviridae/genética , Animais , Divisão Celular , Vírus Defeituosos/genética , Feto , Expressão Gênica , Terapia Genética , Vetores Genéticos , Humanos , Imuno-Histoquímica , Proteína Antagonista do Receptor de Interleucina 1 , Óperon Lac , Pulmão/citologia , Pulmão/enzimologia , Doenças Respiratórias/genética , Doenças Respiratórias/terapia , Ovinos/embriologia , Sialoglicoproteínas/genética , Sialoglicoproteínas/metabolismo , beta-Galactosidase/metabolismo
19.
Am J Physiol ; 268(4 Pt 1): L657-65, 1995 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-7733306

RESUMO

The cystic fibrosis (CF) transmembrane conductance regulator has been localized to both submucosal glands and surface epithelium, suggesting that both glandular and surface epithelium may be important targets for gene therapy. To determine the distribution and efficiency of recombinant adenovirus-mediated gene transfer to human airway submucosal glands, an in vivo model was developed by heterotopically transplanting human bronchial segments from both normal and CF lung tissue into severe combined immunodeficient mice. A serotype 5 E1-deleted recombinant adenovirus containing a lacZ reporter gene driven by the cytomegalovirus promoter (H5.010CMVlacZ) was used to infect the xenografts. Transgene expression was correlated with three factors: 1) viral titer, 2) penetration of microspheres, and 3) dwell time of the viral instillate. At viral titers ranging from 10(8) to 10(11) plaque forming units/ml, expression of the lacZ gene was observed in a subpopulation of epithelial cells within approximately 40% of submucosal glands, with more efficient gene transfer to the ducts than the secretory tubules. Within individual glands, gene transfer varied from < 1% to approximately 20% of submucosal cells, including duct, mucous, and serous cells. Adenovirus-sized fluorescent microspheres were found to penetrate only some of the submucosal glands, suggesting that the gene transfer efficiency to submucosal tubules is due to limited viral particle penetration rather than tropism. Gene transfer to surface epithelial cells was inefficient. However, the percentage of transduced surface epithelial cells increased from < 1% to 5-10% as the dwell time of viral solution was increased from 5 min to 1 h, indicating that the time allowed for virus binding and entry is important for gene transfer efficiency.(ABSTRACT TRUNCATED AT 250 WORDS)


Assuntos
Adenoviridae/fisiologia , Brônquios/fisiologia , Técnicas de Transferência de Genes , Pulmão/fisiologia , Plexo Submucoso/fisiologia , Transplante de Tecidos , Acetilcisteína/farmacologia , Animais , Fibrose Cística/genética , Fluorescência , Vetores Genéticos , Humanos , Imuno-Histoquímica , Camundongos , Camundongos SCID , Microesferas , Transplante Heterólogo
20.
Am J Respir Cell Mol Biol ; 12(2): 142-8, 1995 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-7865213

RESUMO

Early experience with recombinant adenoviruses for gene transfer to airway epithelium suggests that these vectors are associated with the development of inflammation. The mechanisms for this are unclear, but previous work has shown that respiratory viruses can cause increased expression of intercellular adhesion molecule-1 (ICAM-1) on airway epithelial cells. We therefore hypothesized that recombinant adenoviruses may induce ICAM-1 expression and thereby facilitate the development of airway inflammation. To address this, primary cultures of human bronchial epithelial cells were examined for ICAM-1 expression by flow cytometry after infection with a serotype 5, E1/E3-deleted recombinant adenovirus containing the Escherichia coli LacZ reporter gene driven by the cytomegalovirus promoter (Ad.CMVlacZ). Compared with control cells, ICAM-1 expression was unchanged after infection with Ad.CMVlacZ, but increased after infection with wild-type adenovirus. Treatment of Ad.CMVlacZ-infected cells with interferon-gamma (IFN) resulted in increased ICAM-1 expression, but to a lower level than that seen in cells treated with IFN alone, indicating that recombinant adenovirus infection blunted IFN-induced up-regulation of ICAM-1. Adhesion of human leukocytes to human bronchial epithelial cells was not increased after Ad.CMVlacZ infection, thereby excluding an ICAM-1-independent increase in leukocyte-epithelial adhesion. The results for ICAM-1 expression were confirmed in vivo, as immunostaining of human bronchial xenografts infected with Ad.CMVlacZ revealed basilar epithelial staining with ICAM-1, but no increased expression on cells expressing beta-galactosidase. This study demonstrates that unlike other respiratory viruses, recombinant E1/E3-deleted adenovirus does not cause increased ICAM-1 expression on human bronchial epithelium in vitro or in vivo nor increased leukocyte adhesion in vitro.


Assuntos
Adenoviridae/genética , Brônquios/imunologia , Técnicas de Transferência de Genes , Molécula 1 de Adesão Intercelular/biossíntese , Animais , Brônquios/transplante , Brônquios/virologia , Adesão Celular , Células Cultivadas , Citomegalovirus/genética , Epitélio/imunologia , Epitélio/virologia , Genes Reporter , Terapia Genética/efeitos adversos , Vetores Genéticos , Humanos , Inflamação/etiologia , Interferon gama/farmacologia , Óperon Lac , Leucócitos/imunologia , Camundongos , Camundongos SCID , Regiões Promotoras Genéticas , Transplante Heterólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA