Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
1.
Front Immunol ; 13: 1023553, 2022.
Article in English | MEDLINE | ID: mdl-36703990

ABSTRACT

Neutrophil extracellular traps contribute to lung injury in cystic fibrosis and asthma, but the mechanisms are poorly understood. We sought to understand the impact of human NETs on barrier function in primary human bronchial epithelial and a human airway epithelial cell line. We demonstrate that NETs disrupt airway epithelial barrier function by decreasing transepithelial electrical resistance and increasing paracellular flux, partially by NET-induced airway cell apoptosis. NETs selectively impact the expression of tight junction genes claudins 4, 8 and 11. Bronchial epithelia exposed to NETs demonstrate visible gaps in E-cadherin staining, a decrease in full-length E-cadherin protein and the appearance of cleaved E-cadherin peptides. Pretreatment of NETs with alpha-1 antitrypsin (A1AT) inhibits NET serine protease activity, limits E-cadherin cleavage, decreases bronchial cell apoptosis and preserves epithelial integrity. In conclusion, NETs disrupt human airway epithelial barrier function through bronchial cell death and degradation of E-cadherin, which are limited by exogenous A1AT.


Subject(s)
Asthma , Extracellular Traps , Humans , Extracellular Traps/metabolism , Asthma/metabolism , Bronchi , Cell Line , Cadherins/metabolism
2.
Clin Exp Allergy ; 46(2): 317-28, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26263242

ABSTRACT

BACKGROUND: Airway epithelial cells (AEC) are increasingly recognized as a major signalling centre in the pathogenesis of allergic asthma. A previous study demonstrated that epithelial growth factor receptor (EGFR) signalling in AEC regulated key features of allergic airway disease. However, it is unclear what mediators are regulated by EGFR signalling in AEC, although the production of the pro-inflammatory cytokine granulocyte-macrophage colony-stimulating factor (GM-CSF) is EGFR dependent in keratinocytes. OBJECTIVES: To determine whether EGFR signalling regulates GM-CSF production by human AEC downstream of the clinically relevant mediators house dust mite (HDM) and interleukin (IL)-17A and in a mouse model of established allergic asthma. METHODS: EGFR inhibitors were used to determine whether EGFR signalling regulates GM-CSF production by cultured human AEC in response to HDM and IL-17A. The roles of EGFR ligands, p38 mitogen-activated protein kinase (MAPK) and tumour necrosis factor-alpha (TNF-α) converting enzyme (TACE) were also assessed. To determine whether EGFR regulates GM-CSF as well as key asthma characteristics in vivo, mice were chronically exposed to HDM to establish allergic airway disease and then treated with the EGFR inhibitor Erlotinib. RESULTS: EGFR inhibition reduced HDM and IL-17A induced GM-CSF production in a dose-dependent manner in cultured human AEC. GM-CSF production also required amphiregulin, p38 MAPK signalling and protease/TACE activity. In mice with established allergic airway disease, EGFR inhibition reduced levels of GM-CSF and TNF-α, as well as airway hyperreactivity, cellular inflammation, smooth muscle thickening and goblet cell metaplasia without changes in IgE and Th1, Th2 and Th17 cytokines. CONCLUSIONS AND CLINICAL RELEVANCE: Results link HDM, IL-17A, amphiregulin, EGFR and GM-CSF in a mechanistic pathway in AEC and demonstrate that EGFR regulates GM-CSF production and the severity of established disease in a clinically relevant asthma model. These results identify the EGFR→GM-CSF axis as a target for therapeutic development.


Subject(s)
Asthma/immunology , ErbB Receptors/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Respiratory Mucosa/immunology , Signal Transduction/immunology , Animals , Asthma/metabolism , Blotting, Western , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , ErbB Receptors/metabolism , Female , Humans , Interleukin-17/immunology , Mice , Pyroglyphidae/immunology , Respiratory Hypersensitivity/immunology , Respiratory Hypersensitivity/metabolism , Respiratory Mucosa/metabolism
3.
Clin Exp Immunol ; 172(3): 455-65, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23600834

ABSTRACT

Granulocyte-macrophage colony-stimulating factor (GM-CSF) autoantibodies are associated with stricturing behaviour in Crohn disease (CD). We hypothesized that CD ileal lamina propria mononuclear cells (LPMC) would produce GM-CSF autoantibodies and peripheral blood (PB) samples would contain GM-CSF neutralizing capacity (NC). Paediatric CD and control PBMC and ileal biopsies or LPMC were isolated and cultured and GM-CSF, immunoglobulin (Ig)G and GM-CSF autoantibodies production were measured by enzyme-linked immunosorbent assay (ELISA). Basal and GM-CSF-primed neutrophil bacterial killing and signal transducer and activator of transcription 5 (STAT5) tyrosine phosphorylation (pSTAT5) were measured by flow cytometry. GM-CSF autoantibodies were enriched within total IgG for LPMC isolated from CD ileal strictures and proximal margins compared to control ileum. Neutrophil bacterial killing was reduced in CD patients compared to controls. Within CD, neutrophil GM-CSF-dependent STAT5 activation and bacterial killing were reduced as GM-CSF autoantibodies increased. GM-CSF stimulation of pSTAT5 did not vary between controls and CD patients in washed PB granulocytes in which serum was removed. However, GM-CSF stimulation of pSTAT5 was reduced in whole PB samples from CD patients. These data were used to calculate the GM-CSF NC. CD patients with GM-CSF NC greater than 25% exhibited a fourfold higher rate of stricturing behaviour and surgery. The likelihood ratio (95% confidence interval) for stricturing behaviour for patients with elevation in both GM-CSF autoantibodies and GM-CSF NC was equal to 5 (2, 11). GM-CSF autoantibodies are produced by LPMC isolated from CD ileal resection specimens and are associated with reduced neutrophil bacterial killing. CD peripheral blood contains GM-CSF NC, which is associated with increased rates of stricturing behaviour.


Subject(s)
Autoantibodies/biosynthesis , Crohn Disease/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Neutrophils/immunology , Adolescent , Antibodies, Neutralizing/biosynthesis , Blood Bactericidal Activity , Case-Control Studies , Child , Child, Preschool , Constriction, Pathologic , Crohn Disease/metabolism , Crohn Disease/pathology , Female , Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Ileum/immunology , Ileum/metabolism , Ileum/pathology , Infant , Male , Neutrophils/metabolism , STAT5 Transcription Factor/metabolism , Staphylococcus aureus/immunology , Young Adult
6.
Eur Respir J ; 33(5): 1220-2, 2009 May.
Article in English | MEDLINE | ID: mdl-19407056

ABSTRACT

Whole lung lavage (WLL) is currently the standard therapy for pulmonary alveolar proteinosis (PAP). Nevertheless, some PAP patients respond poorly to WLL or require it frequently. The present paper reports a patient with autoimmune PAP with persistent disease despite three WLL treatments over 10 months. Plasmapheresis with ten 1.5-L plasma exchanges was performed, which lowered the serum granulocyte-macrophage colony-stimulating factor (GM-CSF) autoantibody level from 250 microg mL(-1) to 156 microg mL(-1) but did not improve respiratory impairment. Further WLL therapy was required and transiently effective. Serum GM-CSF autoantibody levels declined progressively, reaching a value of 56 microg mL(-1) 80 weeks after completion of plasmapheresis. However, this decrease was not accompanied by clinical improvement and the patient required additional WLL therapy. The results confirm that minor reductions in serum granulocyte-macrophage colony-stimulating factor autoantibody levels from plasmapheresis are not reflected in clinical improvement in the severity of lung disease in pulmonary alveolar proteinosis.


Subject(s)
Plasmapheresis , Pulmonary Alveolar Proteinosis/therapy , Adult , Bronchoalveolar Lavage , Humans , Male , Pulmonary Alveolar Proteinosis/diagnosis
7.
Blood ; 98(9): 2845-52, 2001 Nov 01.
Article in English | MEDLINE | ID: mdl-11675359

ABSTRACT

Macrophage colony-stimulating factor (M-CSF) is one of several hematologic growth factors capable of regulating the survival, proliferation, and differentiation of macrophages, but its role in modulation of the accumulation and function of alveolar macrophages (AMs) in vivo is not well defined. Osteopetrotic (Op/Op) mice have no detectable M-CSF and show variable tissue-specific reductions in macrophage numbers. It was hypothesized that AMs would be decreased in number and have altered function in Op/Op mice because of the absence of M-CSF. Lung macrophages identified by Mac-3 staining in lung sections were decreased in number in 20-day-old Op/Op mice (P <.001) but not Op/Op mice older than 4 months (P =.68) compared with findings in age-matched littermate controls. The numbers of AMs recovered by bronchoalveolar lavage (BAL) were also reduced in young but not adult Op/Op mice compared with controls. Expression of interleukin-3 (IL-3) was increased in the lungs of Op/Op mice compared with controls as determined by quantification of IL-3 cytokine levels (P =.04), bioactivity (P =.02), and messenger RNA transcript levels. AMs of Op/Op mice spontaneously released higher levels of matrix metalloproteinases (MMPs) than AMs of controls as determined by immunohistochemical staining of AMs and zymographic assessment of BAL fluid and AM lysates. Consistent with an increased release of MMP, Op/Op mice had abnormal elastin deposition and spontaneously developed emphysema in the absence of molecular or cellular evidence of lung inflammation. These data show that the AM deficiency observed in young Op/Op mice is spontaneously corrected with age and is associated with increased lung levels of IL-3, spontaneous MMP expression by AMs, and destruction of lung tissue.


Subject(s)
Macrophage Colony-Stimulating Factor/physiology , Macrophages, Alveolar/enzymology , Macrophages, Alveolar/pathology , Matrix Metalloproteinases/metabolism , Osteopetrosis/pathology , Age Factors , Animals , Emphysema/enzymology , Emphysema/etiology , Emphysema/pathology , Interleukin-3/metabolism , Lung/enzymology , Lung/metabolism , Lung/pathology , Macrophage Colony-Stimulating Factor/deficiency , Macrophage Colony-Stimulating Factor/pharmacology , Macrophages, Alveolar/drug effects , Matrix Metalloproteinases/adverse effects , Mice , Mice, Mutant Strains , Osteopetrosis/genetics
8.
Immunity ; 15(4): 557-67, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11672538

ABSTRACT

GM-CSF gene targeted (GM(-/-)) mice are susceptible to respiratory infections and develop alveolar proteinosis due to defects in innate immune function and surfactant catabolism in alveolar macrophages (AMs), respectively. Reduced cell adhesion, phagocytosis, pathogen killing, mannose- and Toll-like receptor expression, and LPS- or peptidoglycan-stimulated TNFalpha release were observed in AMs from GM(-/-) mice. The transcription factor PU.1 was markedly reduced in AMs of GM(-/-) mice in vivo and was restored by selective expression of GM-CSF in the lungs of SPC-GM/GM(-/-) transgenic mice. Retrovirus-mediated expression of PU.1 in AMs from GM(-/-) mice rescued host defense functions and surfactant catabolism by AMs. We conclude that PU.1 mediates GM-CSF-dependent effects on terminal differentiation of AMs regulating innate immune functions and surfactant catabolism by AMs.


Subject(s)
Drosophila Proteins , Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Lung/immunology , Macrophages, Alveolar/immunology , Proto-Oncogene Proteins/physiology , Trans-Activators/physiology , Animals , Cell Adhesion , Cell Differentiation , Cells, Cultured , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Lung/cytology , Macrophages, Alveolar/cytology , Macrophages, Alveolar/microbiology , Membrane Glycoproteins/metabolism , Mice , Mice, Knockout , Models, Biological , Phagocytosis , Proto-Oncogene Proteins/genetics , Pulmonary Surfactants/metabolism , RNA, Messenger/biosynthesis , Receptors, Cell Surface/biosynthesis , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Signal Transduction , Toll-Like Receptors , Trans-Activators/genetics , Transfection
9.
Am J Physiol Lung Cell Mol Physiol ; 280(3): L503-11, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11159034

ABSTRACT

Host inflammatory and immune responses limit viral gene expression after administration of replication-deficient adenoviruses to the lung. The current study asks whether inducible nitric oxide synthase (iNOS) expression and peroxynitrite generation accompanied the inflammatory response following intratracheal administration of adenovirus. Pulmonary iNOS mRNA and protein were increased 2, 7, and 14 days following administration of 2 x 10(9) plaque-forming units of recombinant adenovirus (Av1Luc1) to BALB/c mice. Adenovirus infection was associated with a marked increase in nitrotyrosine staining. Intense nitrotyrosine staining was observed in alveolar macrophages, respiratory epithelial cells, conducting airways, and alveolar spaces 2 days postinfection. Two weeks after exposure to adenovirus, nitrotyrosine staining was detected within alveolar macrophages, suggesting adenovirus enhanced the nitration of proteins that were subsequently taken up by alveolar macrophages. Western blot analysis using anti-nitrotyrosine antibody did not demonstrate accumulation of nitrated surfactant protein A (SP-A), although a small fraction of aggregated SP-A comigrated with a nitrotyrosine-positive protein. iNOS expression, peroxynitrite, and nitrotyrosine generation accompany and may contribute to inflammatory responses to adenovirus in the lung.


Subject(s)
Adenoviridae Infections/metabolism , Lung/metabolism , Nitrates/metabolism , Nitric Oxide Synthase/biosynthesis , Tyrosine/analogs & derivatives , Adenoviridae Infections/pathology , Animals , Blotting, Western , Bronchoalveolar Lavage Fluid/chemistry , Female , Immunohistochemistry , Lung/pathology , Male , Mice , Mice, Inbred BALB C , Nitric Oxide Synthase Type II , Pulmonary Surfactants/metabolism , Tyrosine/metabolism
10.
J Virol ; 74(20): 9655-67, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11000238

ABSTRACT

Adenovirus is a common respiratory pathogen which causes a broad range of distinct clinical syndromes and has recently received attention for its potential for in vivo gene delivery. Although adenovirus respiratory tract infection (ARTI) results in dose-dependent, local inflammation, the pathogenesis of this remains unclear. We hypothesized that alveolar macrophages (AMphi) rapidly internalize adenovirus following in vivo pulmonary administration and then initiate inflammatory signaling within the lung. To evaluate the role of AMphi in the induction of lung inflammation during ARTI in vivo, we directly assessed adenovirus uptake by murine AMphi and correlated uptake with the initiation of proinflammatory gene expression. Stimulation of cytokine (tumor necrosis factor alpha [TNF-alpha], interleukin-6 [IL-6], macrophage inflammatory protein-2 [MIP-2], and MIP-1alpha) expression in the lung was evaluated at the level of mRNA (by reverse transcription-PCR [RT-PCR]) and protein (by enzyme-linked immunosorbent assay) and by identification of cells expressing TNF-alpha and IL-6 mRNA in lung tissues (by in situ hybridization) and isolated lung lavage cells (by RT-PCR). Adenovirus, labeled with the fluorescent dye (Cy3), was rapidly and widely distributed on epithelial surfaces of airways and alveoli and was very rapidly ( approximately 1 min) localized within AMphi. At 30 min after infection AMphi but not airway epithelial or vascular endothelial cells expressed mRNA for TNF-alpha and IL-6, thus identifying AMphi as the cell source of initial cytokine signaling. IL-6, TNF-alpha, MIP-2, and MIP-1alpha levels progressively increased in bronchoalveolar lavage fluid after pulmonary adenovirus infection, and all were significantly elevated at 6 h (P < 0.05). To begin to define the molecular mechanism(s) by which adenovirus initiates the inflammatory signaling in macrophages, TNF-alpha expression from adenovirus-infected RAW264.7 macrophages was evaluated in vitro. TNF-alpha expression was readily detected in adenovirus-infected RAW cell supernatant with kinetics similar to AMphi during in vivo infection. Blockage of virus uptake at specific cellular sites, including internalization (by wortmannin), endosome acidification and/or lysis (by chloroquine) or by Ca(2+) chelation (by BAPTA) completely blocked TNF-alpha expression. In conclusion, results showed that during ARTI, (i) AMphi rapidly internalized adenovirus, (ii) expression of inflammatory mediators was initiated within AMphi and not airway epithelial or other cells, and (iii) the initiation of inflammatory signaling was linked to virion uptake by macrophages occurring at a point after vesicle acidification. These results have implications for our understanding of the role of the AMphi in the initiation of inflammation following adenovirus infection and adenovirus-mediated gene transfer to the lung.


Subject(s)
Adenoviridae Infections/virology , Adenoviridae/physiology , Inflammation/etiology , Macrophages, Alveolar/virology , Respiratory Tract Infections/virology , Acute Disease , Animals , Calcium/physiology , Cell Line , Cytokines/genetics , Gene Transfer Techniques , Genetic Vectors , Mice , Mice, Inbred BALB C , RNA, Messenger/analysis , Tumor Necrosis Factor-alpha/physiology , Virion/isolation & purification
11.
Am J Physiol ; 277(3): L580-8, 1999 09.
Article in English | MEDLINE | ID: mdl-10484466

ABSTRACT

Surfactant protein A (SP-A) is a member of the collectin family of host defense molecules expressed primarily in the epithelial cells of the lung. To determine the role of SP-A in pulmonary adenoviral infection, SP-A-deficient (SP-A -/-) mice were intratracheally infected with a replication-deficient recombinant adenovirus, Av1Luc1. Lung inflammation was markedly increased in SP-A -/- compared with SP-A +/+ mice and was associated with increased hemorrhage and epithelial cell injury. Polymorphonuclear cells in bronchoalveolar lavage fluid (BALF) were increased in SP-A -/- mice after administration of adenovirus. Coadministration of adenovirus and purified human SP-A ameliorated adenoviral-induced lung inflammation in SP-A -/- mice. Concentrations of tumor necrosis factor-alpha (TNF-alpha), interleukin (IL)-6, and IL-1beta were increased in BALF of SP-A -/- mice. Likewise, TNF-alpha, IL-6, macrophage inflammatory protein (MIP)-1alpha, monocyte chemotactic protein-1, and MIP-2 mRNAs were increased in lung homogenates from SP-A -/- mice 6 and 24 h after viral administration. Clearance of adenoviral DNA from the lung and uptake of fluorescent-labeled adenovirus by alveolar macrophages were decreased in SP-A -/- mice. SP-A enhances viral clearance and inhibits lung inflammation during pulmonary adenoviral infection, providing support for the importance of SP-A in antiviral host defense.


Subject(s)
Adenoviridae Infections/virology , Adenoviridae/isolation & purification , Lung/virology , Pneumonia/prevention & control , Proteolipids/physiology , Pulmonary Surfactants/physiology , Adenoviridae Infections/metabolism , Adenoviridae Infections/pathology , Adenoviridae Infections/physiopathology , Animals , Bronchoalveolar Lavage Fluid/cytology , Cytokines/metabolism , Humans , Lung/metabolism , Lung/pathology , Lung/physiopathology , Macrophages, Alveolar/physiology , Mice , Mice, Inbred Strains , Mice, Knockout/genetics , Pneumonia/pathology , Proteolipids/genetics , Proteolipids/pharmacology , Pulmonary Surfactant-Associated Protein A , Pulmonary Surfactant-Associated Proteins , Pulmonary Surfactants/genetics , Pulmonary Surfactants/pharmacology
12.
Am J Respir Cell Mol Biol ; 21(3): 388-94, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10460756

ABSTRACT

In an earlier study, we showed that a recombinant adenovirus vector with deletions in the E1 and E3 regions of the viral genome (AV1LacZ4) induces expression of interleukin (IL)-8 in A549 cells (a human respiratory cell line). IL-8 can be induced through several pathways, including activation by IL-1. We tested the hypothesis that the induction of IL-8 by the AV1LacZ4 adenovirus is accomplished by means of the IL-1/IL-8 activation pathway, which could be blocked by IL-1 receptor antagonist (IRAP). Viral infections of A549 cells were performed at a multiplicity of infection (MOI) of 50 in the presence and absence of IRAP (50 ng/ml). A549 cells were also stimulated with tumor necrosis factor (TNF)-alpha (100 ng/ml), a known stimulant of IL-8, in the presence and absence of IRAP. IL-8 expression was evaluated by Northern blot analysis and enzyme-linked immunosorbent assay. Levels of IL-8 protein and messenger RNA (mRNA) were greater in the infected cells than in the uninfected ones at 24, 48, and 96 h (P < 0.01). Virus-infected cells treated with IRAP expressed 75% less IL-8 mRNA and protein (P < 0.01) than did untreated cells, whereas IRAP pretreatment of TNF-alpha-stimulated cells did not affect IL-8 production. IL-1 production by the virus-infected cells was detectable by concentration of the supernatants and reverse transcription-polymerase chain reaction. We conclude that IL-8 is produced by virus vector-infected cells, partly through IL-1 activation that can be downregulated by IRAP.


Subject(s)
Adenoviridae/genetics , Bronchi/drug effects , Interleukin-8/metabolism , Receptors, Interleukin-1/antagonists & inhibitors , Sialoglycoproteins/pharmacology , Defective Viruses/genetics , Dose-Response Relationship, Drug , Down-Regulation , Epithelial Cells/drug effects , Genetic Vectors , Humans , Interleukin 1 Receptor Antagonist Protein , Interleukin-1/pharmacology , Interleukin-1/physiology , Recombinant Proteins/metabolism , Recombination, Genetic , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Tumor Cells, Cultured
13.
Gene Ther ; 6(1): 98-106, 1999 Jan.
Article in English | MEDLINE | ID: mdl-10341881

ABSTRACT

Fetal gene therapy may prove useful in treating diseases that manifest in the perinatal or early postnatal period. Adenoviruses effectively transfer gene expression to a variety of tissues but also stimulate inflammatory and immune responses. The purpose of this study was to test the hypothesis that exposure of fetal sheep to a first generation adenovirus vector encoding bacterial beta-galactosidase, Av1nBg, before the development of the immune system, is safe, minimizes inflammatory and immune responses and induces tolerance. A total of 22 fetal sheep was studied; of these, two were born with respiratory distress, seven were electively killed and 13 died in utero. The incidence of mortality was higher than the < or = 10% we have experienced with other fetal sheep studies and was not likely related to complications arising from surgical or anesthetic procedures. Inflammatory and fibrotic responses were observed in the lungs and may represent untoward long-term consequences of in utero adenoviral gene therapy. Tolerance to Av1nBg was not established, and repeated exposure to Av1nBg before birth was associated with significant pathology and mortality.


Subject(s)
Adenoviridae/genetics , Fetal Diseases/therapy , Gene Transfer Techniques/adverse effects , Genetic Therapy/adverse effects , Genetic Vectors/adverse effects , Lung Diseases/etiology , Animals , Female , Fetal Death/etiology , Fetal Diseases/immunology , Genetic Therapy/methods , Genetic Vectors/genetics , Gestational Age , Inflammation , Injections , Lung/embryology , Lung/immunology , Lung/pathology , Lung Diseases/immunology , Lung Diseases/pathology , Pregnancy , Sheep
14.
Clin Cardiol ; 22(1 Suppl 1): I23-9, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9929764

ABSTRACT

Adenoviral vectors are promising agents for a number of in vivo gene therapy applications including diseases of the heart and coronary vessels. Efficient intravascular gene transfer to specific sites has been achieved in occluded vessels, but otherwise is hampered by the effect of blood flow on localized vector uptake in the vessel wall. An alternative delivery approach to coronary arteries is the expression of diffusible gene products into the pericardial space surrounding the heart and coronary arteries. However, in vivo pericardial access is comparatively difficult and has been limited to surgical approaches. We hypothesized that efficient adenovirus-mediated gene expression in pericardial lining mesothelium could be achieved by transmyocardial vector delivery to the pericardium. To evaluate this concept, a hollow, helical-tipped penetrating catheter was used to deliver vector-containing fluid directly into the intrapericardial space. The catheter was introduced percutaneously in anesthetized mongrel dogs, advanced into the right ventricle, and the tip passed through the apical right ventricular myocardium under direct radiographic visualization until the open end of the catheter tip resided in the intrapericardial space. Adenoviral vectors expressing either nuclear-localizing beta-galactosidase, cytoplasmic luciferase, or secreted human alpha 1AT reporters (Av1nBg, Av1Lu, or Av1Aa, respectively) were instilled through the catheter into the intrapericardial space. Three days later the animals were sacrificed and reporter gene expression was evaluated in pericardium, epicardium, and multiple other tissues. In animals receiving Av1nBg, beta-galactosidase activity was evident in most of the pericardial lining endothelium, up to 100% in many areas. In animals receiving Av1Lu, luciferase reporter activity was abundant in pericardial tissues, but near-background levels were observed in other organs. In animals receiving Av1Aa, human alpha 1AT was abundant (16-29 mg/ml) in pericardial fluid, but was undetectable in serum. All animals tolerated the procedure well with no electrocardiographic changes and no clinical sequelae. These observations demonstrate highly efficient adenovirus vector delivery and gene transfer and expression in the pericardium and support the feasibility of localized gene therapy via catheter-based pericardial approaches. We suggest that the pericardial sac may serve as a sustained-release protein delivery system for the generation of desired gene products or their metabolites for diffusion into the epicardial region.


Subject(s)
Adenoviridae/genetics , Catheterization/methods , Gene Transfer Techniques , Genetic Vectors , Pericardium , Animals , Catheterization/instrumentation , Coronary Disease/therapy , Diffusion , Dogs , Epithelium , Exudates and Transudates/enzymology , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Viral , Genes, Reporter/genetics , Genetic Therapy , Heart Diseases/therapy , Humans , Injections, Intra-Arterial , Luciferases/genetics , Pericardial Window Techniques , Pericardium/enzymology , Radiography, Interventional , Tissue Distribution , alpha 1-Antitrypsin/genetics , beta-Galactosidase/genetics
15.
Hum Gene Ther ; 9(15): 2207-22, 1998 Oct 10.
Article in English | MEDLINE | ID: mdl-9794205

ABSTRACT

Replication-deficient adenovirus vectors (Avs) have shown high-efficiency gene transfer in a variety of animal models, but demonstrated lower than expected efficiency in the intensely inflammatory milieu of the respiratory tract of individuals with cystic fibrosis (CF). Specific acquired immune responses directed at adenovirus capsid proteins are known to limit the duration of transgene expression and the effectiveness of vector readministration. In these models, however, nonspecific inflammation is also frequently noted to accompany specific immune responses. Because inflammation can occur early after Av administration, we hypothesized that inflammation may block Av-mediated gene transfer in the lung independent of specific immune responses. To evaluate this hypothesis, we measured pulmonary gene transfer and expression in the absence or presence of the potent antiinflammatory agent dexamethasone. To address and eliminate concerns over the potentially confounding effects of systemic, vector-specific acquired immune responses, evaluations were confined to a 3-day period following Av administration and were carried out, in parallel, in normal and immunodeficient (athymic) mice. Dexamethasone significantly reduced Av-associated inflammation in all animals as measured by a significant reduction of blinded, quantitative lung histopathology scores and by reduced proinflammatory cytokine release. Concomitant with reduced inflammation, gene transfer efficiency was significantly increased in both normal and immunodeficient animals as measured by transgene product activity (beta-galactosidase) in total lung homogenates 3 days after vector administration. This finding could not be explained by a direct effect of dexamethasone on transgene specific activity. To begin to understand the molecular mechanisms of Av-induced inflammatory responses, lung levels of the chemoattractive chemokines MIP-2, MIP-1alpha, and MCP-1 were quantified. All were elevated significantly in Av-exposed animals. Dexamethasone reduced levels of MCP-1 and MIP-1alpha, but not MIP-2, consistent with the observed pattern of inflammatory cell changes. Expression of several proinflammatory cytokines including TNF-alpha, IL-6, IL-1beta, and IFN-gamma were also elevated in Av-exposed animals and modulated by dexamethasone. These observations demonstrate that nonspecific inflammation is an important determinant of the efficiency of in vivo pulmonary gene transfer and expression independent of specific immune responses and may have important implications for human gene therapy for diseases of the lung.


Subject(s)
Adenoviridae/genetics , Gene Transfer Techniques , Genetic Vectors , Inflammation , Lung/pathology , Adenoviridae/enzymology , Adenoviridae/immunology , Animals , Anti-Inflammatory Agents/pharmacology , Bronchoalveolar Lavage Fluid/cytology , Cells, Cultured , Chemokines/metabolism , Cytokines/metabolism , Dexamethasone/pharmacology , Epithelial Cells , Gene Expression , Humans , Lung/enzymology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
16.
Hum Gene Ther ; 9(13): 1885-98, 1998 Sep 01.
Article in English | MEDLINE | ID: mdl-9741427

ABSTRACT

Herein, we report that the adenovirus E3-14.7K protein inhibits the inflammatory response to adenovirus in transgenic mice in which the E3-14.7K gene was selectively expressed in the respiratory epithelium, using the human surfactant protein C (SP-C) promoter. E3-14.7K mRNA and protein were detected specifically in the lungs of SPC/E3-14.7K transgenic mice. Responses of the transgenic mice to Av1Luc1, an E1-E3-deleted Ad vector encoding the luciferase reporter gene, were examined, including vector transgene expression and lung inflammation. In wild-type mice, luciferase activity declined rapidly and was lost 14 days following Av1Luc1 administration. The loss of luciferase activity was associated with pulmonary infiltration by macrophages and lymphocytes. In heterozygous SPC/E3-14.7K mice, luciferase activity was increased by 7 days compared with control littermates, and pulmonary infiltration by macrophages was decreased. In homozygous (+/+) SPC/E3-14.7K mice, luciferase activity was increased 7, 14, and 21 days following administration compared with wild-type mice, and lung inflammation was markedly reduced. After Av1Luc1 administration, PCNA staining of bronchiolar and alveolar respiratory epithelial cells was decreased in SPC/E3-14.7K transgenic mice, indicating decreased epithelial cell proliferation, a finding consistent with the observed reduction in inflammation. CD4 and CD8 lymphocyte populations were only mildly altered, while humoral responses to adenoviral vectors were unchanged in the SPC/E3-14.7K mice. The E3-14.7K protein expressed selectively in respiratory epithelial cells suppresses Ad-induced pulmonary epithelial cell cytotoxicity and lung inflammation in vivo and prolongs reporter gene expression.


Subject(s)
Adenoviridae/genetics , Adenovirus E3 Proteins/genetics , Genetic Vectors/genetics , Lung/immunology , Adenoviridae/immunology , Adenovirus E3 Proteins/metabolism , Animals , Antibodies, Viral/blood , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Epithelium/metabolism , Gene Expression Regulation/genetics , Genetic Vectors/immunology , Humans , Luciferases/metabolism , Lung/metabolism , Lung/pathology , Macrophages, Alveolar , Mice , Mice, Transgenic , Organ Specificity , Promoter Regions, Genetic/genetics , Proteolipids/genetics , Pulmonary Surfactants/genetics , RNA, Messenger/analysis , Recombinant Fusion Proteins , Transgenes
17.
Blood ; 91(9): 3273-81, 1998 May 01.
Article in English | MEDLINE | ID: mdl-9558383

ABSTRACT

Hemophilia A is caused by a deficiency of blood coagulation factor VIII (FVIII) and has been widely discussed as a candidate for gene therapy. While the natural canine model of hemophilia A has been valuable for the development of FVIII pharmaceutical products, the use of hemophiliac dogs for gene therapy studies has several limitations such as expense and the long canine generation time. The recent creation of two strains of FVIII-deficient mice provides the first small animal model of hemophilia A. Treatment of hemophiliac mice of both genotypes with potent, human FVIII-encoding adenoviral vectors resulted in expression of biologically active human FVIII at levels, which declined, but remained above the human therapeutic range for over 9 months. The duration of expression and FVIII plasma levels achieved were similar in both hemophiliac mouse strains. Treated mice readily survived tail clipping with minimal blood loss, thus showing phenotypic correction of murine hemophilia A by in vivo gene therapy.


Subject(s)
Factor VIII/genetics , Genetic Therapy/methods , Hemophilia A/therapy , Adenoviridae/genetics , Animals , Antibodies/analysis , Factor VIII/immunology , Factor VIII/metabolism , Gene Expression , Genetic Vectors , Humans , Mice , Mice, Knockout , Recombinant Proteins , Time Factors
18.
Hum Gene Ther ; 8(11): 1331-44, 1997 Jul 20.
Article in English | MEDLINE | ID: mdl-9295128

ABSTRACT

Although replication-deficient adenoviruses efficiently transfer genes into epithelial cells of the lung, host immune responses limit the extent and duration of gene expression. To define further the role of inflammatory responses to first-generation, recombinant, deltaE1, deltaE3 adenovirus in lung pathology and surfactant protein homeostasis, expression of the surfactant proteins SP-A, SP-B, and proSP-C was determined by immunohistochemistry 2, 7, and 14 days following intratracheal administration of 2 x 10(9) pfu of a recombinant adenovirus, Av1Luc1, to BALB/c nu/nu and BALB/c wild-type mice. Two to 7 days after virus administration, an acute inflammatory response was observed in both mouse strains. Respiratory epithelial cells were sloughed, and extracellular accumulation of SP-A and SP-B was detected in the airways. Diminished immunostaining for SP-A and SP-B was noted in type II cells, and SP-A and SP-B mRNA expression was decreased in focal regions of the lungs from both mouse strains. One week after virus administration, immunostaining for proSP-C was markedly increased in cells lining the regenerating alveolar epithelial surfaces. Two weeks after Av1Luc1 treatment of nu/nu mice, immunostaining for SP-A, SP-B, and proSP-C was similar to those patterns observed prior to adenoviral administration. In immunocompetent wild-type mice, however, immunostaining for surfactant proteins was absent in areas associated with chronic lymphocytic infiltration. The recombinant adenoviral vector, Av1Luc1, caused acute inflammatory responses in the respiratory epithelium with disruption of surfactant protein homeostasis in both wild-type and nu/nu mice. Alterations in surfactant homeostasis persisted in wild-type mice. Thus, both acute and thymic-dependent immune responses limit transgene expression and disrupt surfactant protein gene expression and homeostasis. Because surfactant proteins are critical to host defense and to the maintenance of alveolar stability following injury, these findings raise concerns regarding both acute and chronic toxicity of first-generation recombinant adenoviral vectors for gene transfer.


Subject(s)
Adenoviridae Infections/metabolism , Adenoviridae , Homeostasis , Lung/physiology , Pulmonary Surfactants/metabolism , Adenoviridae Infections/complications , Animals , Genetic Vectors/adverse effects , Genetic Vectors/pharmacology , Inflammation/physiopathology , Lung/virology , Mice , Mice, Inbred BALB C , Mice, Nude , Proteolipids/genetics , Proteolipids/metabolism , Pulmonary Surfactant-Associated Protein A , Pulmonary Surfactant-Associated Proteins , Pulmonary Surfactants/genetics , RNA, Messenger/metabolism
19.
Hum Gene Ther ; 8(8): 935-41, 1997 May 20.
Article in English | MEDLINE | ID: mdl-9195216

ABSTRACT

Replication-deficient delta E1a-E3 adenovirus mediates efficient gene transfer to the mouse lung; however it induces a host immune response mediated, in part, by T cells. This immune response is associated with loss of transgene expression. Monoclonal antibodies (mAb) against the T cell receptor (TCR) complex can inhibit both CD4+ and CD8+ T cell responses in vivo and are the most potent anti-T cell agents in clinical use. To determine whether such mAbs can be used to prolong adenovirus-mediated transgene expression, the vector Av1Luc1 (delta E1a-E3 recombinant adenovirus encoding the firefly luciferase gene) was administered intratracheally to C57BL/6 mice on day 0. Three days prior to adenovirus administration (day -3), mice were treated with a single i.p. injection of the anti-TCR mAb H57. Controls received phosphate-buffered saline. Animals were sacrificed on days 3, 14, 28, and 56 and lungs were assessed for transgene expression and histopathology. Luciferase activity decreased markedly in the controls by day 14, but was maintained at high levels in animals receiving anti-TCR mAb. A mild, focal, predominantly neutrophilic inflammation was observed in the alveoli of all mice 3 days after virus administration. In PBS-treated controls, this inflammation progressed to a moderate to severe multifocal, perivascular and peribronchiolar lymphoid infiltration at 14 days. B cells and T cells were present in approximately equal numbers, with CD4+ T cells predominating over CD8+ T cells by 3- to 28-fold. Treatment with H57 resulted in near-complete prevention of the lymphocytic inflammatory infiltrate and increased luciferase activity throughout the 56-day study period, in association with TCR modulation and T cell depletion. Thus, anti-TCR mAb decreases inflammation and prolongs gene expression following adenovirus-mediated gene transfer.


Subject(s)
Adenoviridae/genetics , Gene Transfer Techniques , Genetic Vectors , Pneumonia/prevention & control , Receptors, Antigen, T-Cell/immunology , Animals , Antibodies, Monoclonal , Antibody Formation , Epithelium/immunology , Female , Intubation, Intratracheal , Luciferases/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Time Factors
20.
Gene Ther ; 4(9): 943-9, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9349431

ABSTRACT

The combination of specific gene targeting technologies with efficient gene delivery systems could provide the means to evaluate the concept of anticancer strategies designed to block expression of potentially rate-limiting tumor promoting factors. Here, we constructed adenoviruses expressing hammerhead-ribozymes targeted to two of these factors, the tyrosine kinase receptor HER-2/neu or the growth factor pleiotrophin (PTN). Adenovirus-mediated transduction of either HER-2/neu- or PTN-targeted ribozymes depleted the respective RNAs and inhibited protein expression significantly in three different human cancer cell lines. This resulted in almost complete abrogation of HER-2/neu- or PTN-dependent cancer-cell proliferation, thus demonstrating the feasibility of this approach as a future cancer gene therapy.


Subject(s)
Adenoviridae , Carrier Proteins/genetics , Cytokines/genetics , Gene Targeting , Genes, erbB-2 , Genetic Vectors , Mitogens/genetics , Cell Division/genetics , Gene Expression , Genetic Therapy/methods , Humans , Neoplasms/therapy , RNA, Catalytic/genetics , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...