Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Int J Nanomedicine ; 13: 6345-6357, 2018.
Article in English | MEDLINE | ID: mdl-30349254

ABSTRACT

BACKGROUND: It has been proposed that many hypersensitivity reactions to nanopharmaceuticals represent complement (C)-activation-related pseudoallergy (CARPA), and that pigs provide a sensitive animal model to study the phenomenon. However, a recent study suggested that pulmonary hypertension, the pivotal symptom of porcine CARPA, is not mediated by C in cases of polystyrene nanoparticle (PS-NP)-induced reactions. GOALS: To characterize PS-NPs and reexamine the contribution of CARPA to their pulmonary reactivity in pigs. STUDY DESIGN: C activation by 200, 500, and 750 nm (diameter) PS-NPs and their opsonization were measured in human and pig sera, respectively, and correlated with hemodynamic effects of the same NPs in pigs in vivo. METHODS: Physicochemical characterization of PS-NPs included size, ζ-potential, cryo-transmission electron microscopy, and hydrophobicity analyses. C activation in human serum was measured by ELISA and opsonization of PS-NPs in pig serum by Western blot and flow cytometry. Pulmonary vasoactivity of PS-NPs was quantified in the porcine CARPA model. RESULTS: PS-NPs are monodisperse, highly hydrophobic spheres with strong negative surface charge. In human serum, they caused size-dependent, significant rises in C3a, Bb, and sC5b-9, but not C4d. Exposure to pig serum led within minutes to deposition of C5b-9 and opsonic iC3b on the NPs, and opsonic iC3b fragments (C3dg, C3d) also appeared in serum. PS-NPs caused major hemodynamic changes in pigs, primarily pulmonary hypertension, on the same time scale (minutes) as iC3b fragmentation and opsonization proceeded. There was significant correlation between C activation by different PS-NPs in human serum and pulmonary hypertension in pigs. CONCLUSION: PS-NPs have extreme surface properties with no relevance to clinically used nanomedicines. They can activate C via the alternative pathway, entailing instantaneous opsonization of NPs in pig serum. Therefore, rather than being solely C-independent reactivity, the mechanism of PS-NP-induced hypersensitivity in pigs may involve C activation. These data are consistent with the "double-hit" concept of nanoparticle-induced hypersensitivity reactions involving both CARPA and C-independent pseudoallergy.


Subject(s)
Complement Activation/immunology , Complement C3b/metabolism , Drug Hypersensitivity/immunology , Immunologic Factors/metabolism , Nanoparticles/administration & dosage , Polystyrenes/chemistry , Pulmonary Circulation/immunology , Animals , Complement Activation/drug effects , Drug Hypersensitivity/drug therapy , Humans , Liposomes/chemistry , Male , Nanoparticles/chemistry , Pulmonary Circulation/drug effects , Surface Properties , Swine
2.
Trends Immunol ; 39(11): 890-899, 2018 11.
Article in English | MEDLINE | ID: mdl-30253910

ABSTRACT

Lung capillaries, best known for vital gas exchange, also contribute to neutrophil margination, a phenomenon resulting in large numbers of pulmonary vascular neutrophils. Importantly, the functional relevance of neutrophil margination is unknown. Recent advances in microscopy have altered our understanding of why neutrophils marginate. Specifically, data show that lung capillaries provide a unique anatomical site for neutrophils to capture bloodstream pathogens, which contrasts the conventional monophagocytic-dominated vascular host defense of the spleen and liver. Moreover, lung capillaries provide an efficient site for direct cell-cell communication required for the induction of apoptosis in aged neutrophils. These new ideas transform our views of the pulmonary circulation as a site for immediate neutrophil-mediated host defense and regulation of their life cycle.


Subject(s)
Lung/blood supply , Microvessels/immunology , Neutrophils/immunology , Animals , Cell Communication , Cell Movement , Humans , Immunity, Innate , Phagocytosis , Pulmonary Circulation/immunology
3.
Respir Res ; 18(1): 64, 2017 04 19.
Article in English | MEDLINE | ID: mdl-28424078

ABSTRACT

BACKGROUND: Interleukin 6 (IL-6) is a predictive factor of poor prognosis in patients with acute respiratory distress syndrome (ARDS). However, its acute pulmonary hemodynamic effects and role in lung injury have not been investigated in a clinically relevant murine model of ARDS. METHODS: We used adult C57Bl6 wild-type (WT) and IL-6 knock-out (IL-6KO) mice. The animals received intravenous recombinant human IL-6 (rHuIL-6) or vehicle followed by intratracheal lipopolysaccharide (LPS) or saline before undergoing low tidal volume mechanical ventilation (MV) for 5 h. Before sacrifice, right ventricular systolic pressure and cardiac output were measured and total pulmonary resistance was calculated. After sacrifice, lung inflammation, edema and injury were assessed with bronchoalveolar lavage (BAL) and histology. In other experiments, right ventricular systolic pressure was recorded during hypoxic challenges in uninjured WT mice pretreated with rHuIL-6 or rHuIL-6 + non-selective nitric oxide synthase inhibitor L-NAME or vehicle. RESULTS: IL-6KO(LPS+MV) mice showed a faster deterioration of lung elastic properties and more severe bronchoalveolar cellular inflammation as compared to WT(LPS+MV). Treatment with rHuIL-6 partially prevented this lung deterioration. Total pulmonary resistance was higher in IL-6KO(LPS+MV) mice and this increase was abolished in rHuIL-6-treated IL-6KO mice. Finally, rHuIL-6 reduced hypoxic pulmonary vasoconstriction in uninjured WT mice, an effect that was abolished by co-treatment with L-NAME. CONCLUSIONS: In a double-hit murine model of ARDS, IL-6 deficient mice experienced more severe bronchoalveolar cellular inflammation as compared to wild-type littermates. Furthermore, IL-6 deficiency caused marked acute pulmonary hypertension, which may be, at least partially, due to vasoactive mechanisms. A dysregulation of nitric oxide synthase may account for this observation, a hypothesis that will need to be investigated in future studies.


Subject(s)
Acute Lung Injury/physiopathology , Anti-Inflammatory Agents/immunology , Blood Flow Velocity/immunology , Interleukin-6/immunology , Pulmonary Artery/immunology , Pulmonary Circulation/immunology , Animals , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
4.
Semin Respir Crit Care Med ; 34(5): 551-9, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24037624

ABSTRACT

Pulmonary hypertension is a devastating, life-threatening disorder with no curative options, characterized by elevated pulmonary vascular resistance and secondary right ventricular failure. Although the etiologies of pulmonary arterial hypertension are multiple and its pathogenesis is complex, there is growing evidence that inflammation, endothelial dysfunction, aberrant vascular wall cell proliferation, as well as mutations in the bone morphogenetic protein receptor type 2 gene play a crucial role in triggering pathological vascular remodeling. The present article outlines the current understanding of this disease from the point of view of pathology and pathobiology.


Subject(s)
Endothelium, Vascular/pathology , Hypertension, Pulmonary/pathology , Lung/pathology , Muscle, Smooth, Vascular/pathology , Pulmonary Artery/pathology , Endothelial Cells , Endothelium, Vascular/immunology , Endothelium, Vascular/physiopathology , Familial Primary Pulmonary Hypertension , Fibrosis , Genetic Predisposition to Disease , Humans , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/physiopathology , Inflammation/physiopathology , Lung/blood supply , Lung/immunology , Pulmonary Artery/immunology , Pulmonary Artery/physiopathology , Pulmonary Circulation/immunology
5.
Eur J Clin Invest ; 42(12): 1342-9, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22984929

ABSTRACT

Neutrophils are the most abundant circulating white cell in humans and play a crucial role in the innate immune response. Accumulation and activation of neutrophils, together with delayed clearance, have been shown to be a key event in the pathogenesis of acute lung injury. Previously, it has been proposed that there is substantial pooling of neutrophils within the pulmonary vasculature, even under physiological conditions, making the lung especially vulnerable to neutrophil-mediated tissue injury. However, more recent evidence suggests that only primed neutrophils accumulate in the pulmonary vasculature. This article examines the evidence for these two opposing views and proposes a new two-step model for the recruitment of neutrophils into the lung. Firstly, neutrophils that become primed, by exposure to a range of inflammatory mediators or physicochemical perturbations, become shape changed and stiff because of alterations in their cytoskeleton, and as a result, accumulate within the pulmonary circulation. In the absence of further stimuli, the healthy pulmonary vasculature is able to selectively retained these primed cells, allow them to 'de-prime' and be released back into the circulation in a quiescent, state. If this pulmonary 'de-priming' mechanism fails, or a second insult occurs, such as ventilator-associated barotrauma, which causes loss of alveolar integrity, primed neutrophils migrate from the pulmonary vasculature into the interstitial space with resultant lung injury. This canonical 'two step' model highlights the importance of neutrophil priming in the genesis of lung injury and the importance of adopting strategies to minimise alveolar injury.


Subject(s)
Acute Lung Injury/etiology , Neutrophil Activation/physiology , Neutrophils/physiology , Pulmonary Circulation/physiology , Acute Lung Injury/immunology , Animals , Humans , Models, Animal , Models, Biological , Neutrophil Activation/immunology , Neutrophils/immunology , Pulmonary Circulation/immunology
6.
Am J Physiol Lung Cell Mol Physiol ; 297(2): L238-50, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19465514

ABSTRACT

Recent studies demonstrate that sustained hypoxia induces the robust accumulation of leukocytes and mesenchymal progenitor cells in pulmonary arteries (PAs). Since the factors orchestrating hypoxia-induced vascular inflammation are not well-defined, the goal of this study was to identify mediators potentially responsible for recruitment to and retention and differentiation of circulating cells within the hypoxic PA. We analyzed mRNA expression of 44 different chemokine/chemokine receptor, cytokine, adhesion, and growth and differentiation genes in PAs obtained via laser capture microdissection in adjacent lung parenchyma and in systemic arteries by RT-PCR at several time points of hypoxic exposure (1, 7, and 28 days) in Wistar-Kyoto rats. Analysis of inflammatory cell accumulation and protein expression of selected genes was concomitantly assessed by immunochemistry. We found that hypoxia induced progressive accumulation of monocytes and dendritic cells in the vessel wall with few T cells and no B cells or neutrophils. Upregulation of stromal cell-derived factor-1 (SDF-1), VEGF, growth-related oncogene protein-alpha (GRO-alpha), C5, ICAM-1, osteopontin (OPN), and transforming growth factor-beta (TGF-beta) preceded mononuclear cell influx. With time, a more complex pattern of gene expression developed with persistent upregulation of adhesion molecules (ICAM-1, VCAM-1, and OPN) and monocyte/fibrocyte growth and differentiation factors (TGF-beta, endothelin-1, and 5-lipoxygenase). On return to normoxia, expression of many genes (including SDF-1, monocyte chemoattractant protein-1, C5, ICAM-1, and TGF-beta) rapidly returned to control levels, changes that preceded the disappearance of monocytes and reversal of vascular remodeling. In conclusion, sustained hypoxia leads to the development of a complex, PA-specific, proinflammatory microenvironment capable of promoting recruitment, retention, and differentiation of circulating monocytic cell populations that contribute to vascular remodeling.


Subject(s)
Hypertension, Pulmonary/immunology , Hypoxia/immunology , Pneumonia/immunology , Pulmonary Artery/immunology , Pulmonary Circulation/immunology , Animals , Aorta, Thoracic/physiology , Cell Adhesion Molecules/genetics , Chemokines/genetics , Chronic Disease , Cytokines/genetics , Dendritic Cells/immunology , Dendritic Cells/pathology , Gene Expression Regulation/immunology , Intercellular Signaling Peptides and Proteins/genetics , Lung/blood supply , Lung/physiology , Monocytes/immunology , Monocytes/pathology , Oligonucleotide Array Sequence Analysis , Pneumonia/pathology , Pulmonary Artery/pathology , Rats , Rats, Inbred WKY , Receptors, Chemokine/genetics
7.
Clin Hemorheol Microcirc ; 41(2): 127-36, 2009.
Article in English | MEDLINE | ID: mdl-19252235

ABSTRACT

Impaired deformability might contribute to the accumulation of activated leukocytes within pulmonary microcapillaries, leading to acute lung injury. The purpose of our study was to investigate changes in leukocyte deformability during periods of inflammation after esophagectomy. The study group comprised 20 patients who underwent esophagectomy. Changes in leukocyte deformability were investigated by examining filtration through a silicon microchannel, which simulated human pulmonary microcapillaries. Changes in the neutrophil cytoskeleton were investigated by measuring neutrophil F-actin assembly. The severity of patient clinical outcome was evaluated by the lung injury score. Leukocyte filtration through the microchannel was significantly weaker in esophagectomy patients than in healthy subjects (p<0.01). After esophagectomy, filtration was further impaired compared with preoperative values (p<0.05). The neutrophil F-actin content was higher in patients than in controls (p<0.01), and increased after esophagectomy compared with preoperative values (p<0.01). We concluded that circulating leukocytes showed reduced deformability and appeared to be sequestered within microcapillaries after esophagectomy. Changes in neutrophil cytoskeleton were considered to be responsible for the reduced deformability. Leukocyte accumulation within pulmonary microcapillaries might be related to the pathogenesis of lung injury after esophagectomy.


Subject(s)
Acute Lung Injury/immunology , Esophagectomy/adverse effects , Neutrophils/physiology , Postoperative Complications/immunology , Acute Lung Injury/physiopathology , Case-Control Studies , Female , Flow Cytometry , Hemorheology/immunology , Humans , Inflammation/physiopathology , Male , Microcirculation/immunology , Microfluidic Analytical Techniques , Neutrophils/ultrastructure , Pulmonary Circulation/immunology
8.
Int Arch Allergy Immunol ; 149(3): 251-8, 2009.
Article in English | MEDLINE | ID: mdl-19218818

ABSTRACT

BACKGROUND: Asthma and allergic airway inflammation are associated with persistent structural alterations in the bronchi, i.e. airway remodeling. Previous studies have shown that during allergic airway inflammation, similar structural alterations may also be evoked in the pulmonary circulation. However, it remained unknown whether remodeling of the pulmonary circulation is as persistent as airway remodeling. The aim of this study is to investigate the reversibility and resolution of vascular remodeling, induced by allergic airway inflammation. METHODS: A validated mouse model of allergic airway inflammation, utilizing ovalbumin as allergen, was employed. Animals were sacrificed 1 day, 1 week or 1 month after the last allergen exposure, and different parameters of remodeling (smooth muscle mass, proliferation of smooth muscle cells and endothelial cells as well as number of myofibroblasts and procollagen-I-producing cells) were investigated and quantified histologically. RESULTS: Allergen exposure resulted in allergic airway inflammation characterized by a transient leukocyte infiltration and in structural alterations in both airway and vascular compartments. The increase in vascular smooth muscle mass and endothelial proliferation persisted at 1 month after the last allergen exposure. The other parameters and cellular inflammatory response returned to baseline within 1 month after the last allergen challenge. CONCLUSIONS: Based on the findings in this study, we conclude that acute allergic airway inflammation, although being initiated from the airways, is able to evoke similar long-term structural alterations in pulmonary vessels as described for bronchi.


Subject(s)
Asthma/pathology , Endothelium, Vascular/pathology , Lung/blood supply , Lung/pathology , Pneumonia/pathology , Pulmonary Circulation/immunology , Allergens/immunology , Animals , Asthma/immunology , Asthma/metabolism , Cell Proliferation , Endothelium, Vascular/immunology , Eosinophilia/immunology , Eosinophilia/pathology , Female , Lung/immunology , Mice , Mice, Inbred BALB C , Myocytes, Smooth Muscle/immunology , Ovalbumin/immunology , Pneumonia/immunology , Procollagen/biosynthesis , Procollagen/immunology
9.
Respir Res ; 9: 69, 2008 Oct 24.
Article in English | MEDLINE | ID: mdl-18950499

ABSTRACT

BACKGROUND: Bile-duct ligated (BDL) rats recruit pulmonary intravascular macrophages (PIMs) and are highly susceptible to endotoxin-induced mortality. The mechanisms of this enhanced susceptibility and mortality in BDL rats, which are used as a model of hepato-pulmonary syndrome, remain unknown. We tested a hypothesis that recruited PIMs promote endotoxin-induced mortality in a rat model. METHODS: Rats were subjected to BDL to induce PIM recruitment followed by treatment with gadolinium chloride (GC) to deplete PIMs. Normal and BDL rats were treated intravenously with E. coli lipopolysaccharide (LPS) with or without GC pre-treatment followed by collection and analyses of lungs for histopathology, electron microscopy and cytokine quantification. RESULTS: BDL rats recruited PIMs without any change in the expression of IL-1beta, TNF-alpha and IL-10. GC caused reduction in PIMs at 48 hours post-treatment (P < 0.05). BDL rats treated intravenously with E. coli LPS died within 3 hours of the challenge while the normal LPS-treated rats were euthanized at 6 hours after the LPS treatment. GC treatment of rats 6 hours or 48 hours before LPS challenge resulted in 80% (1/5) and 100% (0/5) survival, respectively, at 6 hours post-LPS treatment. Lungs from BDL+LPS rats showed large areas of perivascular hemorrhages compared to those pre-treated with GC. Concentrations of IL-1beta, TNF-alpha and IL-10 were increased in lungs of BDL+LPS rats compared to BDL rats treated with GC 48 hours but not 6 hours before LPS (P < 0.05). CONCLUSION: We conclude that PIMs increase susceptibility for LPS-induced lung injury and mortality in this model, which is blocked by a reduction in their numbers or their inactivation.


Subject(s)
Disease Models, Animal , Lung/immunology , Lung/pathology , Macrophages/immunology , Pneumonia/immunology , Pulmonary Circulation/immunology , Animals , Lipopolysaccharides , Lung/drug effects , Macrophages/drug effects , Male , Pneumonia/chemically induced , Pneumonia/pathology , Rats , Rats, Sprague-Dawley , Survival , Survival Rate
10.
Blood ; 109(4): 1593-601, 2007 Feb 15.
Article in English | MEDLINE | ID: mdl-17032919

ABSTRACT

Plasminogen activator inhibitor type-1 (PAI-1) levels are consistently elevated in patients with severe pneumonia and sepsis and highly predictive for an unfavorable outcome. In addition, pneumonia is associated with strongly elevated PAI-1 levels in the pulmonary compartment. However, whether PAI-1 causally affects antibacterial host defense in vivo remains unknown. We report here that pneumonia caused by the common respiratory pathogen Klebsiella pneumoniae is associated with local production of PAI-1 in the lungs of wild-type mice. PAI-1 deficiency impaired host defense as reflected by enhanced lethality and increased bacterial growth and dissemination in mice with a targeted deletion of the PAI-1 gene. Conversely, transgenic overexpression of PAI-1 in the lung using a replication-defective adenoviral vector markedly improved host defense against Klebsiella pneumonia and sepsis. PAI-1 deficiency reduced accumulation of neutrophils in the lungs during pneumonia, whereas PAI-1 overexpression in healthy lungs resulted in neutrophil influx, suggesting that PAI-1 protects the host against Klebsiella pneumonia by promoting neutrophil recruitment to the pulmonary compartment. These data demonstrate for the first time that PAI-1 is essential for host defense against severe Gram-negative pneumonia.


Subject(s)
Klebsiella pneumoniae , Plasminogen Activator Inhibitor 1/immunology , Pneumonia/immunology , Animals , Gram-Negative Bacteria , Klebsiella pneumoniae/immunology , Lung/immunology , Lung/microbiology , Mice , Mice, Knockout , Neutrophils/immunology , Plasminogen Activator Inhibitor 1/blood , Plasminogen Activator Inhibitor 1/deficiency , Pneumonia/microbiology , Pneumonia/pathology , Prognosis , Pulmonary Circulation/immunology , Sepsis
11.
Environ Health Perspect ; 114(9): 1354-60, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16966088

ABSTRACT

Ambient air is polluted with a mixture of pulmonary toxicants. Previous studies indicate that prior exposure to atmospheric oxidant pollutants such as ozone may significantly alter the response to other pollutants, such as 1-nitronaphthalene (1-NN) . 1-NN, a component of the particulate exhaust from diesel engines, has been found at low concentrations in ambient air. Using a metabolomic approach, we investigated inflammatory responses in arachidonic and linoleic acid biochemical cascades (35 metabolites) and the expression of 19 cytokines/chemokines at three time points (2, 6, and 24 hr) following exposure to 1-NN with and without prior long-term O3 exposure. Long-term O3 exposure is associated with biochemical changes that have been shown to render the lung resistant to further O3 exposure. This study indicates that airways of O3-tolerant rats exhibited a low level of chronic inflammation, rendering the lungs more susceptible to other environmental pollutants such as 1-NN. Specifically, a 12.5-mg/kg dose of 1-NN to O3-tolerant rats produced significantly higher levels of cysteinyl-leukotrienes in bronchiolar lavage fluid even when compared to a 50-mg/kg dose of 1-NN in rats exposed to filtered air. Collectively, these results indicate that the combination of exposures as encountered in polluted ambient air are considerably more injurious to the lung than would be anticipated from previous studies employing single exposures. The observed synergism between O3 and 1-NN may be causally related to a shift in a T-helper 1 to T-helper 2 immune response in the airways.


Subject(s)
Air Pollutants/toxicity , Inflammation Mediators/metabolism , Inhalation Exposure , Lung/drug effects , Naphthalenes/toxicity , Ozone/toxicity , Pulmonary Circulation/drug effects , Administration, Inhalation , Animals , Chemokines/immunology , Chemokines/metabolism , Cytokines/immunology , Cytokines/metabolism , Dose-Response Relationship, Drug , Inflammation Mediators/immunology , Lung/immunology , Lung/physiology , Male , Pulmonary Circulation/immunology , Pulmonary Circulation/physiology , Rats , Rats, Sprague-Dawley , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism
12.
J Appl Physiol (1985) ; 98(2): 715-21, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15649883

ABSTRACT

Data are rapidly accumulating in support of the idea that circulating monocytes and/or mononuclear fibrocytes are recruited to the pulmonary circulation of chronically hypoxic animals and that these cells play an important role in the pulmonary hypertensive process. Hypoxic induction of monocyte chemoattractant protein-1, stromal cell-derived factor-1, vascular endothelial growth factor-A, endothelin-1, and tumor growth factor-beta(1) in pulmonary vessel wall cells, either directly or indirectly via signals from hypoxic lung epithelial cells, may be a critical first step in the recruitment of circulating leukocytes to the pulmonary circulation. In addition, hypoxic stress appears to induce release of increased numbers of monocytic progenitor cells from the bone marrow, and these cells may have upregulated expression of receptors for the chemokines produced by the lung circulation, which thus facilitates their specific recruitment to the pulmonary site. Once present, macrophages/fibrocytes may exert paracrine effects on resident pulmonary vessel wall cells stimulating proliferation, phenotypic modulation, and migration of resident fibroblasts and smooth muscle cells. They may also contribute directly to the remodeling process through increased production of collagen and/or differentiation into myofibroblasts. In addition, they could play a critical role in initiating and/or supporting neovascularization of the pulmonary artery vasa vasorum. The expanded vasa network may then act as a conduit for further delivery of circulating mononuclear cells to the pulmonary arterial wall, creating a feedforward loop of pathological remodeling. Future studies will need to determine the mechanisms that selectively induce leukocyte/fibrocyte recruitment to the lung circulation under hypoxic conditions, their direct role in the remodeling process via production of extracellular matrix and/or differentiation into myofibroblasts, their impact on the phenotype of resident smooth muscle cells and adventitial fibroblasts, and their role in the neovascularization observed in hypoxic pulmonary hypertension.


Subject(s)
Hypertension, Pulmonary/immunology , Hypoxia/immunology , Leukocytes/immunology , Lung/immunology , Lymphocyte Activation/immunology , Pulmonary Circulation/immunology , Animals , Humans , Hypertension, Pulmonary/etiology , Hypoxia/complications , Models, Immunological
13.
Am J Physiol Lung Cell Mol Physiol ; 287(2): L307-17, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15075248

ABSTRACT

Murine Mycoplasma pulmonis infection induces chronic lung and airway inflammation accompanied by profound and persistent microvascular remodeling in tracheobronchial mucosa. Because matrix metalloproteinase (MMP)-2 and -9 are important for angiogenesis associated with placental and long bone development and skin cancer, we hypothesized that they contribute to microvascular remodeling in airways infected with M. pulmonis. To test this hypothesis, we compared microvascular changes in airways after M. pulmonis infection of wild-type FVB/N mice with those of MMP-9(-/-) and MMP-2(-/-)/MMP-9(-/-) double-null mice and mice treated with the broad-spectrum MMP inhibitor AG3340 (Prinomastat). Using zymography and immunohistochemistry, we find that MMP-2 and MMP-9 rise strikingly in lungs and airways of infected wild-type FVB/N and C57BL/6 mice, with no zymographic activity or immunoreactivity in MMP-2(-/-)/MMP-9(-/-) animals. However, microvascular remodeling as assessed by Lycopersicon esculentum lectin staining of whole-mounted tracheae is as severe in infected MMP-9(-/-), MMP-2(-/-)/MMP-9(-/-) and AG3340-treated mice as in wild-type mice. Furthermore, all groups of infected mice develop similar inflammatory infiltrates and exhibit similar overall disease severity as indicated by decrease in body weight and increase in lung weight. Uninfected wild-type tracheae show negligible MMP-2 immunoreactivity, with scant MMP-9 immunoreactivity in and around growing cartilage. By contrast, MMP-2 appears in epithelial cells of infected, wild-type tracheae, and MMP-9 localizes to a large population of infiltrating leukocytes. We conclude that despite major increases in expression, MMP-2 and MMP-9 are not essential for microvascular remodeling in M. pulmonis-induced chronic airway inflammation.


Subject(s)
Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/genetics , Neovascularization, Pathologic/physiopathology , Pneumonia, Mycoplasma/physiopathology , Animals , Epithelial Cells/enzymology , Gene Expression Regulation, Enzymologic/immunology , Immunohistochemistry , Leukocytes/enzymology , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/pathology , Pneumonia, Mycoplasma/immunology , Pneumonia, Mycoplasma/pathology , Pulmonary Circulation/immunology , Trachea/enzymology , Trachea/immunology , Trachea/pathology
14.
Blood ; 103(2): 639-47, 2004 Jan 15.
Article in English | MEDLINE | ID: mdl-14504080

ABSTRACT

Asthma is associated with airway remodeling. Evidence of platelet recruitment to the lungs of asthmatics after allergen exposure suggests platelets participate in various aspects of asthma; although their importance is unknown in the context of airway remodeling, their involvement in atherosclerosis is established. Studies from our laboratory have shown a requirement for platelets in pulmonary leukocyte recruitment in a murine model of allergic lung inflammation. Presently, the effects of platelet depletion and corticosteroid administration on airway remodeling and lung function were examined. Ovalbumin (OVA)-sensitized mice, exposed to aerosolized OVA for 8 weeks, demonstrated epithelial and smooth muscle thickening, and subepithelial reticular fiber deposition in the distal airways. The depletion of platelets via an immunologic (antiplatelet antisera) or nonimmunologic (busulfan) method, markedly reduced airway remodeling. In contrast, dexamethasone administration did not affect epithelial thickening or subepithelial fibrosis, despite significantly inhibiting leukocyte recruitment. Thus, pathways leading to certain aspects of airway remodeling may not depend on leukocyte recruitment, whereas platelet activation is obligatory. OVA-sensitized mice exhibited airway hyperresponsiveness (AHR) compared with sham-sensitized mice following chronic OVA exposure. Neither platelet depletion nor dexamethasone administration inhibited chronic AHR; thus, mechanisms other than inflammation and airway remodeling may be involved in the pathogenesis of chronic AHR.


Subject(s)
Asthma/immunology , Blood Platelets/physiology , Hypersensitivity/immunology , Respiratory Mucosa/immunology , Adrenal Cortex Hormones/pharmacology , Animals , Asthma/pathology , Bronchoalveolar Lavage Fluid , Dexamethasone/pharmacology , Disease Models, Animal , Hypersensitivity/pathology , Immunoglobulin E/blood , Inflammation/immunology , Inflammation/pathology , Leukocytes/immunology , Mice , Mice, Inbred C57BL , Ovalbumin/immunology , Platelet Count , Pulmonary Circulation/immunology , Pulmonary Circulation/physiology , Respiratory Function Tests , Respiratory Mucosa/drug effects , Respiratory Mucosa/pathology , Thrombocytopenia/immunology
15.
Am J Physiol Lung Cell Mol Physiol ; 285(3): L654-63, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12794008

ABSTRACT

The interaction of circulating leukocytes with lung microvessels is a critical event in the recruitment of effector cells into the interstitial tissue during episodes of inflammation, including smoking-induced chronic airway disease. In the present study, murine lung tissue transplanted into a dorsal skinfold window chamber in nude mice was used as a model system to study nicotine-induced leukocyte trafficking in vivo. The revascularized lung microvessels were determined to be of pulmonary origin based on their ability to constrict in response to hypoxia. We demonstrated that nicotine significantly enhanced rolling and adhesion of leukocytes within lung microvessels comprising arterioles and postcapillary venules in a dose-dependent manner, but failed to induce leukocyte emigration. Nicotine-induced rolling and adhesion was significantly higher in venules than in arterioles. Treatment of mice with monoclonal antibodies (MAbs) against L-, E-, or P-selectin after exposure of lung allografts to nicotine resulted in variable but significant inhibition of nicotine-induced rolling, whereas nicotine-induced subsequent adhesion was inhibited by MAbs against L- and P-selectin but not E-selectin. Exposure of lung allografts to nicotine along with PD-98059, a mitogen-activated protein kinase (MAPK)-specific inhibitor, resulted in significant inhibition of nicotine-induced rolling and adhesion. In vitro, exposure of murine lung endothelial cells to nicotine resulted in increased phosphorylation of mitogen-activated/extracellular signal-regulated protein kinase 1/2, which could be blocked by PD-98059. Overall, these results suggest that nicotine-induced inflammation in the airways could potentially be due to MAPK-mediated, selectin-dependent leukocyte-endothelial cell interactions in the lung microcirculation.


Subject(s)
Endothelium, Vascular/drug effects , Leukocyte Rolling/drug effects , Lung Transplantation , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Selectins/metabolism , Animals , Antibodies/pharmacology , Cell Adhesion/immunology , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Leukocyte Rolling/immunology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/immunology , Mice , Mice, Inbred BALB C , Mice, Nude , Microcirculation/immunology , Pulmonary Circulation/immunology , Selectins/immunology
16.
Am J Physiol Lung Cell Mol Physiol ; 285(1): L232-9, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12730078

ABSTRACT

Epinephrine (Epi) increases lymphocyte traffic to lung. We investigated whether Epi also modulates pulmonary cell-mediated immune responses in vivo. C57BL/6 mice were immunized with hen-egg lysozyme (HEL) on day 0, challenged with HEL intratracheally at day 12, and killed at day 15. Mice received Epi (0.5 mg/kg) subcutaneously during the sensitization phase, days 1-7 (Epi-SP), or the effector phase, days 12-14 (Epi-EP); controls received saline subcutaneously. Epi-SP mice showed increased airway inflammation (P < 0.03) and pulmonary angiitis (P < 0.04) characterized by endothelialitis and subendothelial fibrin deposition. Macrophages and granulocytes were increased in perivascular cuffs in situ (P < 0.001). CD3+ lymphocytes increased in the bronchoalveolar lavage fluid, whereas NK1.1+ and CD4+CD25+ lymphocytes decreased (all P < 0.05). Atenolol, a selective beta1-adrenoreceptor (AR) antagonist, inhibited the increased vascular and airway inflammation and the reduction in CD4+CD25+ lymphocytes (all P < 0.05) yielded by Epi, whereas all alpha/beta-AR blockers inhibited airway inflammation. We conclude that Epi-EP selectively promotes vascular inflammation in vivo via a beta1-receptor-mediated mechanism.


Subject(s)
Adrenergic beta-Agonists/pharmacology , Epinephrine/pharmacology , Pulmonary Circulation/immunology , Receptors, Adrenergic, beta-1/metabolism , Vasculitis/immunology , Animals , CD11b Antigen/analysis , Chemokines/genetics , Cortisone/metabolism , Female , Gene Expression/drug effects , Gene Expression/immunology , Granulocytes/immunology , Lymphocyte Subsets/immunology , Mice , Mice, Inbred C57BL , Monocytes/chemistry , Monocytes/immunology , Pneumonia/chemically induced , Pneumonia/immunology , Pulmonary Circulation/drug effects , Receptors, Adrenergic, beta-1/immunology , Specific Pathogen-Free Organisms , Vasculitis/chemically induced
17.
Xenotransplantation ; 10(2): 120-31, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12588645

ABSTRACT

BACKGROUND: The role of complement in hyperacute lung xenograft rejection has not been elucidated. The present study evaluates the effect of complement (C) C3/C5 convertase inhibition on hyperacute rejection of pig lung by human blood. METHODS: In an established ex-vivo model, lungs from pigs heterozygous for human decay accelerating factor (hDAF), non-transgenic littermate control pigs, or farm-bred pigs were perfused with fresh human blood that was either unmodified or treated with soluble complement receptor type 1 (sCR1: TP10, 100 microg/ml). RESULTS: Non-transgenic lungs from littermate controls had a median survival time of 35 min (range 5 to 210; P = 0.25 vs. farm-bred piglets: median 5 min, range 5 to 10). Lungs expressing hDAF survived for a median of 90 min (range 10 to 161; P = 0.5 and 0.01 vs. littermate and farm-bred controls, respectively), with sCR1, whereas hDAF (-) lungs failed by 35 min (range 6 to 307), hDAF (+) lungs survived for 330 min (range 39 to 577) [P = 0.002 vs. farm-bred; P = 0.08 vs. hDAF (-); P = 0.17 vs. sCR1/hDAF (-)]. The rise in pulmonary vascular resistance (PVR) at 5 min was blunted only by hDAF (+) with sCR1 (0.26 +/- 0.2 vs. 0.5 to 0.7 mmHg/ml/min for other groups). Plasma C3a and sC5b-9 and tissue deposition of C5b-9 were dramatically diminished using sCR1, and further decreased in association with hDAF. Histamine and thromboxane were produced rapidly in all groups. CONCLUSION: Complement plays an important role in lung HAR. However, even potent inhibition of C3/C5 convertase, both membrane bound in lung and by a soluble-phase inhibitor in the blood, does not prevent activation of inflammatory responses known to be particularly injurious to the lung. Our findings implicate a role for innate immune pathways resistant to efficient complement regulation. The role of anti-species antibody, coagulation pathway dysregulation, and additional environmental or genetic influences remain to be defined.


Subject(s)
Complement C3-C5 Convertases/antagonists & inhibitors , Graft Rejection/immunology , Lung Transplantation/immunology , Transplantation, Heterologous/immunology , Acute Disease , Animals , Animals, Genetically Modified , Antibodies, Heterophile/blood , Blood Cell Count , CD55 Antigens/genetics , Cell Membrane/metabolism , Complement C3-C5 Convertases/metabolism , Complement System Proteins/immunology , Complement System Proteins/metabolism , Graft Rejection/metabolism , Graft Rejection/mortality , Graft Survival/immunology , Histamine/metabolism , Humans , Pulmonary Circulation/immunology , Receptors, Complement/metabolism , Survival Rate , Swine , Thromboxanes/metabolism , Vascular Resistance/immunology
18.
Ann Clin Lab Sci ; 32(2): 164-70, 2002.
Article in English | MEDLINE | ID: mdl-12017199

ABSTRACT

The purpose of this study was to examine the effects of growth hormone (GH) on nuclear factor kappa B (NF-kappaB) activation and organ injury induced by lipopolysaccharide (LPS) in rats. Male Wistar rats were divided into 6 groups treated with saline, LPS (5 mg/kg), LPS plus GH (0.5, 1.0, 2.0 mg/kg), or GH (2.0 mg/kg) alone for 2 or 4 hr. NF-kappaB activity and I-kappaB level in lung, lung accumulation of neutrophils, and lung microvascular injury were measured. LPS-challenged rats had increased NF-kappaB activity and decreased I-kappaB level in lung, compared to controls. GH dramatically enhanced NF-kappaB activation and I-kappaB degradation induced by LPS challenge. LPS plus GH treatment increased lung accumulation of neutrophils, compared with LPS treatment. Also, subsequently, GH treatment increased lung microvascular injury induced by LPS. These findings suggest that treatment with GH is harmful, instead of beneficial, to LPS-induced organ injury. Increased NF-kappaB activation may be a critical in vivo mechanism that mediates GH action on LPS-induced organ injury. Thus, it is appropriate to rethink GH administration in critical illnesses; further studies are required to evaluate the safety and clinical benefits of GH administration in such conditions.


Subject(s)
Growth Hormone/pharmacology , I-kappa B Proteins , Lung Diseases/drug therapy , Lung Diseases/immunology , NF-kappa B/metabolism , Pulmonary Circulation/immunology , Animals , Capillary Permeability/drug effects , Capillary Permeability/immunology , DNA-Binding Proteins/metabolism , Lipopolysaccharides , Lung Diseases/etiology , Male , Microcirculation/drug effects , Microcirculation/immunology , Neutrophils/immunology , Peritonitis/chemically induced , Peritonitis/complications , Peritonitis/immunology , Pulmonary Circulation/drug effects , Rats , Rats, Wistar , Sepsis/chemically induced , Sepsis/complications , Sepsis/immunology
20.
Am J Physiol Lung Cell Mol Physiol ; 282(5): L959-67, 2002 May.
Article in English | MEDLINE | ID: mdl-11943660

ABSTRACT

Because of its relative inaccessibility, inflammatory cell extravasation within the airway circulation in vivo has been difficult to investigate in real time. A new method has been established using intravital microscopy in the anesthetized rat to visualize leukocytes in superficial postcapillary venules of the trachea. This technique has been validated using local superfusion of lipopolysaccharide (LPS) and N-formyl-methionyl-leucyl-phenylalanine (FMLP). Basal leukocyte rolling velocity (55.4 +/- 9.3 microm/s) and adhesion (1.4 +/- 0.3 cells/100 microm) were monitored in postcapillary venules (33.9 +/- 1.3 microm diameter). At all time points up to 90 min, these parameters were unaltered in control rats (n = 7). In contrast, vessels exposed to 1 microg/ml of LPS (n = 6) exhibited a 57% reduction in leukocyte rolling velocity and an increase in the number of adherent cells (4.7 +/- 1 cells/100 microm, P < 0.05). Superfusion with 0.1 microM of FMLP (n = 6) also resulted in a 45% reduction in rolling velocity and an increase in adherent cells (4 +/- 0.7 cells/100 microm, P < 0.05). Histological evaluation confirmed local stimulus-induced leukocyte extravasation. These results demonstrate leukocyte recruitment in the airway microvasculature and provide an important new method to study airway inflammation in real time.


Subject(s)
Neutrophils/cytology , Pulmonary Circulation/immunology , Trachea/blood supply , Trachea/immunology , Animals , Cell Adhesion/drug effects , Cell Adhesion/immunology , Leukocyte Count , Lipopolysaccharides/pharmacology , Male , Microcirculation/immunology , Microscopy/instrumentation , Microscopy/methods , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Pneumonia/chemically induced , Pneumonia/immunology , Rats , Rats, Sprague-Dawley , Venules/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...