Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters











Publication year range
1.
J Immunol ; 208(6): 1467-1482, 2022 03 15.
Article in English | MEDLINE | ID: mdl-35173037

ABSTRACT

Asthma is a chronic disease of childhood, but for unknown reasons, disease activity sometimes subsides as children mature. In this study, we present clinical and animal model evidence suggesting that the age dependency of childhood asthma stems from an evolving host response to respiratory viral infection. Using clinical data, we show that societal suppression of respiratory virus transmission during coronavirus disease 2019 lockdown disrupted the traditional age gradient in pediatric asthma exacerbations, connecting the phenomenon of asthma remission to virus exposure. In mice, we show that asthmatic lung pathology triggered by Sendai virus (SeV) or influenza A virus is highly age-sensitive: robust in juvenile mice (4-6 wk old) but attenuated in mature mice (>3 mo old). Interestingly, allergen induction of the same asthmatic traits was less dependent on chronological age than viruses. Age-specific responses to SeV included a juvenile bias toward type 2 airway inflammation that emerged early in infection, whereas mature mice exhibited a more restricted bronchiolar distribution of infection that produced a distinct type 2 low inflammatory cytokine profile. In the basal state, aging produced changes to lung leukocyte burden, including the number and transcriptional landscape of alveolar macrophages (AMs). Importantly, depleting AMs in mature mice restored post-SeV pathology to juvenile levels. Thus, aging influences chronic outcomes of respiratory viral infection through regulation of the AM compartment and type 2 inflammatory responses to viruses. Our data provide insight into how asthma remission might develop in children.


Subject(s)
Age Factors , Aging/physiology , Asthma/immunology , COVID-19/immunology , Influenza A virus/physiology , Influenza, Human/immunology , Lung/immunology , Orthomyxoviridae Infections/immunology , Respirovirus Infections/immunology , SARS-CoV-2/physiology , Sendai virus/physiology , Th2 Cells/immunology , Animals , Asthma/epidemiology , COVID-19/epidemiology , Cytokines/metabolism , Humans , Influenza, Human/epidemiology , Mice , Mice, Inbred C57BL , United States/epidemiology
2.
Sci Rep ; 10(1): 9027, 2020 06 03.
Article in English | MEDLINE | ID: mdl-32493985

ABSTRACT

Encouraging clinical results using immune checkpoint therapies to target the PD-1 axis in a variety of cancer types have paved the way for new immune therapy trials in brain tumor patients. However, the molecular mechanisms that regulate expression of the PD-1 pathway ligands, PD-L1 and PD-L2, remain poorly understood. To address this, we explored the cell-intrinsic mechanisms of constitutive PD-L1 and PD-L2 expression in brain tumors. PD-L1 and PD-L2 expression was assessed by flow cytometry and qRT-PCR in brain tumor cell lines and patient tumor-derived brain tumor-initiating cells (BTICs). Immunologic effects of PD-L2 overexpression were evaluated by IFN-γ ELISPOT. CD274 and PDCD1LG2 cis-regulatory regions were cloned from genomic DNA and assessed in full or by mutating and/or deleting regulatory elements by luciferase assays. Correlations between clinical responses and PD-L1 and PD-L2 expression status were evaluated in TCGA datasets in LGG and GBM patients. We found that a subset of brain tumor cell lines and BTICs expressed high constitutive levels of PD-L1 and PD-L2 and that PD-L2 overexpression inhibited neoantigen specific T cell IFN-γ production. Characterization of novel cis-regulatory regions in CD274 and PDCD1LG2 lead us to identify that GATA2 is sufficient to drive PD-L1 and PD-L2 expression and is necessary for PD-L2 expression. Importantly, in TCGA datasets, PD-L2 correlated with worse clinical outcomes in glioma patients.. By perturbing GATA2 biology, targeted therapies may be useful to decrease inhibitory effects of PD-L2 in the microenvironment.


Subject(s)
B7-H1 Antigen/biosynthesis , Brain Neoplasms/immunology , GATA2 Transcription Factor/metabolism , Glioma/immunology , Programmed Cell Death 1 Ligand 2 Protein/biosynthesis , Animals , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , B7-H1 Antigen/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , GATA2 Transcription Factor/genetics , Glioma/genetics , Glioma/metabolism , Humans , Mice , Programmed Cell Death 1 Ligand 2 Protein/genetics , Programmed Cell Death 1 Ligand 2 Protein/immunology , Programmed Cell Death 1 Ligand 2 Protein/metabolism , Tumor Microenvironment
3.
PLoS One ; 7(9): e45546, 2012.
Article in English | MEDLINE | ID: mdl-23029085

ABSTRACT

Laminin-332 is a heterotrimeric basement membrane component comprised of the α3, ß3, and γ2 laminin chains. Laminin-332 modulates epithelial cell processes, such as adhesion, migration, and differentiation and is prominent in many embryonic and adult tissues. In skin, laminin-332 is secreted by keratinocytes and is a key component of hemidesmosomes connecting the keratinocytes to the underlying dermis. In mice, lack of expression of any of the three Laminin-332 chains result in impaired anchorage and detachment of the epidermis, similar to that seen in human junctional epidermolysis bullosa, and death occurs within a few days after birth. To bypass the early lethality of laminin-332 deficiency caused by the knockout of the mouse laminin γ2 chain, we expressed a dox-controllable human laminin γ2 transgene under a keratinocyte-specific promoter on the laminin γ2 (Lamc2) knockout background. These mice appear similar to their wild-type littermates, do not develop skin blisters, are fertile, and survive >1.5 years. Immunofluorescence analyses of the skin showed that human laminin γ2 colocalized with mouse laminin α3 and ß3 in the basement membrane zone underlying the epidermis. Furthermore, the presence of "humanized" laminin-332 in the epidermal basement membrane zone rescued the alterations in the deposition of hemidesmosomal components, such as plectin, collagen type XVII/BP180, and integrin α6 and ß4 chains, seen in conventional Lamc2 knockout mice, leading to restored formation of hemidesmosomes. These mice will be a valuable tool for studies of organs deficient in laminin-332 and the role of laminin-332 in skin, including wound healing.


Subject(s)
Blister/genetics , Gene Expression , Genes, Lethal , Keratinocytes/metabolism , Laminin/genetics , Alleles , Animals , Basement Membrane/metabolism , Blister/pathology , Blister/prevention & control , Epidermal Cells , Epidermis/metabolism , Gene Order , Gene Targeting , Hemidesmosomes/metabolism , Hemidesmosomes/ultrastructure , Humans , Laminin/metabolism , Mice , Mice, Knockout , Mouth Mucosa/metabolism , Mouth Mucosa/pathology , Phenotype , Protein Binding , Protein Transport , Skin/metabolism , Skin/pathology , Transgenes
4.
Am J Respir Crit Care Med ; 183(7): 876-84, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21057003

ABSTRACT

RATIONALE: Matrix metalloprotease (MMP)-9 is an elastolytic endopeptidase produced by activated macrophages that may be involved in the development of human pulmonary emphysema and could be inhibited with existing compounds. Mouse models have demonstrated that excess MMP-9 production can result in permanent alveolar destruction. OBJECTIVES: To determine if MMP-9 causes cigarette smoke-induced emphysema using MMP-9 knockout mice and human samples. METHODS: Mouse lungs were analyzed for inflammation and airspace enlargement using a mainstream smoke-exposure model. Human macrophage mRNA was isolated from subjects with emphysema by laser capture microdissection. Human blood monocyte mRNA was isolated from subjects with greater than 30 pack-year smoking history. Human gene expression was determined by quantitative polymerase chain reaction and compared with emphysema severity determined by automated computed tomography analysis. Plasma Clara cell secretory protein and surfactant protein-D were quantified to measure ongoing lung injury. MEASUREMENTS AND MAIN RESULTS: Mice deficient in MMP-9 develop the same degree of cigarette smoke-induced inflammation and airspace enlargement as strain-matched controls. Macrophages are the predominant source of MMP-9 production in human emphysema specimens and similar quantities of macrophage MMP-9 mRNA is present in areas of lung with and without emphysema. Circulating monocytes produce more MMP-9 in individuals with advanced emphysema severity despite no correlation of MMP-9 with markers of ongoing lung damage. CONCLUSIONS: These results suggest that MMP-9 in humans who smoke is similar to smoke-exposed mice, where MMP-9 is present in emphysematous lung but not correlated with the emphysema. To the degree that the mechanisms of emphysema in humans who smoke resemble the mouse model, these data suggest specific inhibition of MMP-9 is unlikely to be an effective therapy for cigarette smoke-induced emphysema. Clinical trial registered with www.clinicaltrials.gov (NCT 00757120).


Subject(s)
Matrix Metalloproteinase 9/metabolism , Pulmonary Emphysema/enzymology , Pulmonary Emphysema/pathology , Pulmonary Surfactant-Associated Protein D/metabolism , Aged , Analysis of Variance , Animals , Biopsy, Needle , Bronchoalveolar Lavage Fluid/cytology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Humans , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/metabolism , Pulmonary Emphysema/chemically induced , RNA, Messenger/analysis , Reference Values , Reverse Transcriptase Polymerase Chain Reaction , Smoke , Smoking , Tissue Culture Techniques
5.
Am J Physiol Lung Cell Mol Physiol ; 296(6): L1051-8, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19329539

ABSTRACT

We investigated the influence of extracellular matrix on transport properties of mouse alveolar epithelial cell (AEC) monolayers (MAECM) and transdifferentiation of isolated mouse alveolar epithelial type II (AT2) cells into an alveolar epithelial type I (AT1) cell-like phenotype. Primary mouse AT2 cells plated on laminin 5-coated polycarbonate filters formed monolayers with transepithelial resistance (R(T)) and equivalent short-circuit current (I(EQ)) of 1.8 kOmega.cm(2) and 5.3 microA/cm(2), respectively, after 8 days in culture. Amiloride (10 microM), ouabain (0.1 mM), and pimozide (10 microM) decreased MAECM I(EQ) to 40%, 10%, and 65% of its initial value, respectively. Sequential addition of pimozide and amiloride, in either order, revealed that their inhibitory effects are additive, suggesting that cyclic nucleotide-gated channels contribute to amiloride-insensitive active ion transport across MAECM. Ussing chamber measurements of unidirectional ion fluxes across MAECM under short-circuit conditions indicated that net absorption of Na(+) in the apical-to-basolateral direction is comparable to net ion flux calculated from the observed short-circuit current: 0.38 and 0.33 microeq.cm(-2).h(-1), respectively. Between days 1 and 9 in culture, AEC demonstrated increased expression of aquaporin-5 protein, an AT1 cell marker, and decreased expression of pro-surfactant protein-C protein, an AT2 cell marker, consistent with transition to an AT1 cell-like phenotype. These results demonstrate that AT1 cell-like MAECM grown on laminin 5-coated polycarbonate filters exhibit active and passive transport properties that likely reflect the properties of intact mouse alveolar epithelium. This mouse in vitro model will enhance the study of AEC derived from mutant strains of mice and help define important structure-function correlations.


Subject(s)
Epithelial Cells/cytology , Epithelial Cells/physiology , Respiratory Mucosa/cytology , Respiratory Mucosa/physiology , Amiloride/pharmacology , Animals , Biological Transport, Active/drug effects , Biological Transport, Active/physiology , Bronchodilator Agents/pharmacology , Cell Culture Techniques , Cells, Cultured , Chlorides/metabolism , Diffusion Chambers, Culture , Dopamine Antagonists/pharmacology , Electric Impedance , Enzyme Inhibitors/pharmacology , Epithelial Cells/drug effects , Mice , Mice, Inbred Strains , Ouabain/pharmacology , Phenotype , Pimozide/pharmacology , Pulmonary Alveoli/cytology , Pulmonary Alveoli/physiology , Sodium/metabolism , Sodium Channel Blockers/pharmacology , Terbutaline/pharmacology
6.
Am J Respir Cell Mol Biol ; 39(4): 400-11, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18441282

ABSTRACT

Cigarette smoke (CS) is the main risk factor for chronic obstructive pulmonary disease (COPD). Terminal bronchioles are critical zones in the pathophysiology of COPD, but little is known about the cellular and molecular changes that occur in cells lining terminal bronchioles in response to CS. We subjected C57BL/6 mice to CS (6 d/wk, up to 6 mo), looked for morphologic changes lining the terminal bronchioles, and used laser capture microdissection to selectively isolate cells in terminal bronchioles to study gene expression. Morphologic and immunohistochemical analyses showed that Clara cell predominance remained despite 6 months of CS exposure. Since Clara cells have a role in protection against oxidative stress, we focused on the expression of antioxidant/detoxification genes using microarray analysis. Of the 35 antioxidant/detoxification genes with at least 2.5-fold increased expression in response to 6 months of CS exposure, 21 were NF-E2-related factor 2 (Nrf2)-regulated genes. Among these were cytochrome P450 1b1, glutathione reductase, thioredoxin reductase, and members of the glutathione S-transferase family, as well as Nrf2 itself. In vitro studies using immortalized murine Clara cells (C22) showed that CS induced the stabilization and nuclear translocation of Nrf2, which correlated with the induction of antioxidant and detoxification genes. Furthermore, decreasing Nrf2 expression by siRNA resulted in a corresponding decrease in CS-induced expression of several antioxidant and detoxification genes by C22 cells. These data suggest that the protective response by Clara cells to CS exposure is predominantly regulated by the transcription factor Nrf2.


Subject(s)
Bronchioles/pathology , NF-E2-Related Factor 2/physiology , Pulmonary Disease, Chronic Obstructive/metabolism , Smoke/adverse effects , Animals , Antioxidants/metabolism , Bronchioles/metabolism , Cell Nucleus/metabolism , Female , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Protein Transport , Pulmonary Disease, Chronic Obstructive/etiology , Pulmonary Disease, Chronic Obstructive/pathology
7.
J Biol Chem ; 283(15): 9513-22, 2008 Apr 11.
Article in English | MEDLINE | ID: mdl-18178964

ABSTRACT

Proteolytic processing of laminin-332 by matrix metalloproteinase (MMP)-2 and MMP-14 has been shown to yield fragments that are promigratory for epithelial cells. During acute and chronic inflammation, proteases are elaborated by neutrophils and macrophages that can degrade basement membranes. We investigated the susceptibility of laminin-332 to degradation by the following neutrophil and macrophage proteases: neutrophil elastase (NE), cathepsin G, proteinase-3, and MMPs-2, -8, -9, and -12. Protease-specific differences were seen in the capacity to cleave the individual chains of laminin-332. NE and MMP-12 showed the greatest activity toward the gamma2 chain, generating a fragment similar in size to the gamma2x fragment generated by MMP-2. The digestion pattern of laminin-332 by degranulated neutrophils was nearly identical to that generated with NE alone. Digestion by supernatants of degranulated neutrophils was blocked by an inhibitor of NE, and NE-deficient neutrophils were essentially unable to digest laminin-332, suggesting that NE is the major neutrophil-derived protease that degrades laminin-332. In vivo, laminin gamma2 fragments were found in the bronchoalveolar lavage fluid of wild-type mice treated with lipopolysaccharide, whereas that obtained from NE-deficient mice showed a different cleavage pattern. In addition, NE cleaved a synthetic peptide derived from the region of human laminin gamma2 containing the MMP-2 cleavage site, suggesting that NE may generate laminin-332 fragments that are also promigratory. Both laminin-332 fragments generated by NE digestion and NE-digested laminin gamma2 peptide were found to be chemotactic for neutrophils. Collectively, these data suggest that degradation of laminin-332 by NE generates fragments with important biological activities.


Subject(s)
Cell Adhesion Molecules/metabolism , Chemotactic Factors/metabolism , Leukocyte Elastase/metabolism , Neutrophils/enzymology , Peptides/metabolism , Animals , Bronchoalveolar Lavage Fluid , Cathepsin G , Cathepsins/genetics , Cathepsins/metabolism , Cell Adhesion Molecules/genetics , Cell Degranulation/physiology , Chemotactic Factors/genetics , Humans , Laminin/genetics , Laminin/metabolism , Leukocyte Elastase/genetics , Lipopolysaccharides/pharmacology , Male , Matrix Metalloproteinases/genetics , Matrix Metalloproteinases/metabolism , Mice , Mice, Knockout , Myeloblastin/genetics , Myeloblastin/metabolism , Neutrophils/cytology , Peptides/genetics , Rats , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Substrate Specificity/physiology , Kalinin
8.
Respir Res ; 9: 1, 2008 Jan 07.
Article in English | MEDLINE | ID: mdl-18179694

ABSTRACT

BACKGROUND: Clara cells are the epithelial progenitor cell of the small airways, a location known to be important in many lung disorders. Although migration of alveolar type II and bronchiolar ciliated epithelial cells has been examined, the migratory response of Clara cells has received little attention. METHODS: Using a modification of existing procedures for Clara cell isolation, we examined mouse Clara cells and a mouse Clara-like cell line (C22) for adhesion to and migration toward matrix substrate gradients, to establish the nature and integrin dependence of migration in Clara cells. RESULTS: We observed that Clara cells adhere preferentially to fibronectin (Fn) and type I collagen (Col I) similar to previous reports. Migration of Clara cells can be directed by a fixed gradient of matrix substrates (haptotaxis). Migration of the C22 cell line was similar to the Clara cells so integrin dependence of migration was evaluated with this cell line. As determined by competition with an RGD containing-peptide, migration of C22 cells toward Fn and laminin (Lm) 511 (formerly laminin 10) was significantly RGD integrin dependent, but migration toward Col I was RGD integrin independent, suggesting that Clara cells utilize different receptors for these different matrices. CONCLUSION: Thus, Clara cells resemble alveolar type II and bronchiolar ciliated epithelial cells by showing integrin mediated pro-migratory changes to extracellular matrix components that are present in tissues after injury.


Subject(s)
Cell Adhesion/physiology , Chemotaxis/physiology , Epithelial Cells/cytology , Epithelial Cells/physiology , Extracellular Matrix Proteins/pharmacology , Extracellular Matrix/physiology , Trachea/cytology , Trachea/physiology , Animals , Cell Adhesion/drug effects , Cells, Cultured , Chemotaxis/drug effects , Mice
9.
Am J Respir Cell Mol Biol ; 38(1): 8-15, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17673686

ABSTRACT

Tumor necrosis factor (TNF)-alpha is a cytokine produced by alveolar macrophages in response to LPS in the lung. Clara cells are bronchiolar epithelial cells that produce a variety of proinflammatory cytokines in response to LPS but not to TNF-alpha. In this study, we examined whether TNF-alpha affects Clara cell cytokine production in the setting of LPS stimulation. Using a transformed murine Clara cell line (C22), we observed that both LPS and TNF-alpha induced production of keratinocyte-derived chemokine (KC) and monocyte chemoattractant protein (MCP)-1. We also found that simultaneous LPS and TNF-alpha stimulation is synergistic for KC production, but additive for MCP-1 production. By using a Transwell coculture system of RAW264.7 macrophages and Clara cells isolated from C57Bl/6 mice, we found that macrophages produce a soluble factor that enhances Clara cell KC production in response to LPS. Cocultures of Clara cells from mice deficient in TNF-alpha receptors with RAW264.7 macrophages demonstrated that the effect of macrophages on Clara cells is mediated primarily via TNF-alpha. To determine whether these findings occur in vivo, we treated wild-type and TNF receptor-deficient mice intratracheally with LPS and examined the expression of KC. LPS-treated, TNF receptor-deficient mice showed much less KC mRNA in airway epithelial cells compared with wild-type mice. In contrast, a similar number of KC-expressing cells was seen in the lung periphery. Thus, upregulation of KC by Clara cells in the setting of LPS stimulation is largely dependent on TNF-alpha originating from alveolar macrophages. These findings shed light on macrophage-Clara cell interactions in regulating the pulmonary inflammatory response to LPS.


Subject(s)
Cell Communication/physiology , Chemokine CXCL1/biosynthesis , Epithelial Cells/metabolism , Lipopolysaccharides/pharmacology , Macrophages, Alveolar/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Animals , Cell Communication/drug effects , Cell Line , Chemokine CCL2/biosynthesis , Chemokine CCL2/genetics , Chemokine CXCL1/genetics , Coculture Techniques , Drug Synergism , Epithelial Cells/cytology , Lipopolysaccharides/agonists , Lung/cytology , Lung/metabolism , Macrophages, Alveolar/cytology , Mice , Mice, Knockout , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/metabolism , Tumor Necrosis Factor-alpha/agonists , Up-Regulation/drug effects , Up-Regulation/physiology
10.
Am J Physiol Lung Cell Mol Physiol ; 293(2): L383-92, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17526599

ABSTRACT

Airway epithelial cells secrete proinflammatory mediators in response to LPS, but cytokine production by a prominent nonciliated bronchiolar epithelial cell, the Clara cell, specifically, is unknown. To investigate Clara cell cytokine production in response to LPS, we used a transformed murine Clara cell line, C22, and isolated Clara cells from C57Bl/6 mice. Stimulation of both cell types with LPS resulted in significant upregulation of keratinocyte-derived chemokine (KC) and monocyte chemoattractant protein-1, but did not induce TNF-alpha production. To determine whether LPS induces cytokine production by Clara cells in vivo, LPS was instilled intratracheally into mice. KC was expressed by Clara cells, alveolar type 2 cells, and alveolar macrophages, 2 h after LPS administration, as determined by in situ hybridization. TNF-alpha, although not expressed in airway epithelial cells, was expressed primarily in alveolar macrophages in response to LPS. To assess the impact of Clara cells on KC and TNF-alpha production in the lung in the early response to LPS, mice were treated with naphthalene to selectively induce Clara cell injury before LPS stimulation. KC expression in the airways and the lung periphery, and KC and TNF-alpha levels in the bronchoalveolar lavage fluid, were significantly reduced in naphthalene-treated vs. vehicle-treated mice after LPS stimulation. Furthermore, transwell cocultures of C22 cells and RAW264.7 macrophages indicated that C22 cells released a soluble factor(s) in response to LPS that enhanced macrophage production of TNF-alpha. These results indicate that Clara cells elaborate cytokines and modulate the lung innate immune response to LPS.


Subject(s)
Lipopolysaccharides/pharmacology , Pneumonia/immunology , Pneumonia/pathology , Respiratory Mucosa/immunology , Respiratory Mucosa/pathology , Animals , Bronchoalveolar Lavage Fluid/immunology , Chemokine CCL2/metabolism , Chemokine CXCL1 , Chemokines, CXC/metabolism , Cytokines/metabolism , Epithelial Cells/immunology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Naphthalenes/toxicity , Respiratory Mucosa/metabolism , Tumor Necrosis Factor-alpha/metabolism
11.
J Clin Invest ; 116(3): 753-9, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16470245

ABSTRACT

Mice lacking macrophage elastase (matrix metalloproteinase-12, or MMP-12) were previously shown to be protected from the development of cigarette smoke-induced emphysema and from the accumulation of lung macrophages normally induced by chronic exposure to cigarette smoke. To determine the basis for macrophage accumulation in experimental emphysema, we now show that bronchoalveolar lavage fluid from WT smoke-exposed animals contained chemotactic activity for monocytes in vitro that was absent in lavage fluid from macrophage elastase-deficient mice. Fractionation of the bronchoalveolar lavage fluid demonstrated the presence of elastin fragments only in the fractions containing chemotactic activity. An mAb against elastin fragments eliminated both the in vitro chemotactic activity and cigarette smoke-induced monocyte recruitment to the lung in vivo. Porcine pancreatic elastase was used to recruit monocytes to the lung and to generate emphysema. Elastin fragment antagonism in this model abrogated both macrophage accumulation and airspace enlargement.


Subject(s)
Elastin/physiology , Emphysema/metabolism , Emphysema/pathology , Peptide Fragments/physiology , Animals , Bronchoalveolar Lavage Fluid , Chemotaxis, Leukocyte , Disease Models, Animal , Disease Progression , Macrophages, Alveolar/metabolism , Matrix Metalloproteinase 12 , Metalloendopeptidases/deficiency , Metalloendopeptidases/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/metabolism , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/pathology , Smoking
12.
Dev Biol ; 282(1): 111-25, 2005 Jun 01.
Article in English | MEDLINE | ID: mdl-15936333

ABSTRACT

Laminin alpha5 is prominent in the basement membrane of alveolar walls, airways, and pleura in developing and adult lung. Targeted deletion of laminin alpha5 in mice causes developmental defects in multiple organs, but embryonic lethality has precluded examination of the latter stages of lung development. To identify roles for laminin alpha5 in lung development, we have generated an inducible lung epithelial cell-specific Lama5 null (SP-CLama5(fl/-)) mouse through use of the Cre/loxP system, the human surfactant protein C promoter, and the reverse tetracycline transactivator. SP-CLama5(fl/-) embryos exposed to doxycycline from E6.5 died a few hours after birth. Compared to control littermates, SP-CLama5(fl/-) lungs had dilated, enlarged distal airspaces, but basement membrane ultrastructure was preserved. Distal epithelial cell differentiation was perturbed, with a marked reduction of alveolar type II cells and a virtual absence of type I cells. Cell proliferation was reduced and apoptosis was increased. Capillary density was diminished, and this was associated with a decrease in total lung VEGF production. Overall, these findings indicate that epithelial laminin alpha5, independent of its structural function, is necessary for murine lung development, and suggest a role for laminin alpha5 in signaling pathways that promote alveolar epithelial cell differentiation and VEGF expression.


Subject(s)
Cell Differentiation/physiology , Epithelial Cells/physiology , Laminin/metabolism , Pulmonary Alveoli/embryology , Signal Transduction/physiology , Vascular Endothelial Growth Factor A/metabolism , Animals , Apoptosis/physiology , Basement Membrane/ultrastructure , Blotting, Western , DNA Primers , Doxycycline , Epithelial Cells/metabolism , Immunohistochemistry , Laminin/genetics , Mice , Mice, Mutant Strains , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Reverse Transcriptase Polymerase Chain Reaction
13.
Am J Pathol ; 166(2): 377-86, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15681822

ABSTRACT

The molecular mechanisms that cause emphysema are complex but most theories suggest that an excess of proteinases is a crucial requirement. This paradigm is exemplified by severe deficiency of the key anti-elastase within the lung: alpha(1)-antitrypsin. The Z mutant of alpha(1)-antitrypsin has a point mutation Glu342Lys in the hinge region of the molecule that renders it prone to intermolecular linkage and loop-sheet polymerization. Polymers of Z alpha(1)-antitrypsin aggregate within the liver leading to juvenile liver cirrhosis and the resultant plasma deficiency predisposes to premature emphysema. We show here that polymeric alpha(1)-anti-trypsin co-localizes with neutrophils in the alveoli of individuals with Z alpha(1)-antitrypsin-related emphysema. The significance of this finding is underscored by the excess of neutrophils in these individuals and the demonstration that polymers cause an influx of neutrophils when instilled into murine lungs. Polymers exert their effect directly on neutrophils rather than via inflammatory cytokines. These data provide an explanation for the accelerated tissue destruction that is characteristic of Z alpha(1)-antitrypsin-related emphysema. The transition of native Z alpha(1)-antitrypsin to polymers inactivates its anti-proteinase function, and also converts it to a proinflammatory stimulus. These findings may also explain the progression of emphysema in some individuals despite alpha(1)-antitrypsin replacement therapy.


Subject(s)
Emphysema/pathology , Neutrophils/metabolism , Polymers/chemistry , Pulmonary Alveoli/metabolism , alpha 1-Antitrypsin/chemistry , Ammonium Sulfate/pharmacology , Animals , Bronchoalveolar Lavage Fluid , Chemotaxis , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Homozygote , Humans , Immunohistochemistry , Leukocyte Elastase/metabolism , Lung/metabolism , Mice , Mice, Inbred C57BL , Models, Biological , Point Mutation
14.
J Immunol ; 174(3): 1621-9, 2005 Feb 01.
Article in English | MEDLINE | ID: mdl-15661925

ABSTRACT

Tissue injury triggers inflammatory responses that may result in release of degradation products or exposure of cryptic domains of extracellular matrix components. Previously, we have shown that a cryptic peptide (AQARSAASKVKVSMKF) in the alpha-chain of laminin-10 (alpha5beta1gamma1), a prominent basement membrane component, is chemotactic for both neutrophils (PMNs) and macrophages (Mphis) and induces matrix metalloproteinase-9 (MMP-9) production. To determine whether AQARSAASKVKVSMKF has additional effects on inflammatory cells, we performed microarray analysis of RNA from RAW264.7 Mphis stimulated with AQARSAASKVKVSMKF. Several cytokines and cytokine receptors were increased >3-fold in response to the laminin alpha5 peptide. Among these were TNF-alpha and one of its receptors, the p75 TNFR (TNFR-II), increasing 3.5- and 5.7-fold, respectively. However, the peptide had no effect on p55 TNFR (TNFR-I) expression. Corroborating the microarray data, the protein levels of TNF-alpha and TNFR-II were increased following stimulation of RAW264.7 cells with AQARSAASKVKVSMKF. In addition, we determined that the production of TNF-alpha and TNFR-II in response to AQARSAASKVKVSMKF preceded the production of MMP-9. Furthermore, using primary Mphis from mice deficient in TNFR-I, TNFR-II, or both TNF-alpha receptors (TNFRs), we determined that AQARSAASKVKVSMKF induces MMP-9 expression by Mphis through a pathway triggered by TNFR-II. However, TNF-alpha signaling is not required for AQARSAASKVKVSMKF-induced PMN release of MMP-9 or PMN emigration. These data suggest that interactions of inflammatory cells with basement membrane components may orchestrate immune responses by inducing expression of cytokines, recruitment of inflammatory cells, and release of proteinases.


Subject(s)
Chemotactic Factors/physiology , Inflammation Mediators/physiology , Laminin/physiology , Peptide Fragments/physiology , Signal Transduction/immunology , Tumor Necrosis Factor-alpha/physiology , Amino Acid Sequence , Animals , Cell Line , Gene Expression Regulation/immunology , Humans , Macrophages, Peritoneal/enzymology , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Matrix Metalloproteinase 9/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Monocytes/immunology , Monocytes/metabolism , Neutrophil Infiltration/genetics , Neutrophil Infiltration/immunology , Oligonucleotide Array Sequence Analysis , Receptors, Tumor Necrosis Factor, Type I/biosynthesis , Receptors, Tumor Necrosis Factor, Type I/deficiency , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type II/biosynthesis , Receptors, Tumor Necrosis Factor, Type II/deficiency , Receptors, Tumor Necrosis Factor, Type II/genetics , Signal Transduction/genetics , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics
15.
Am J Pathol ; 163(6): 2329-35, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14633606

ABSTRACT

To address the role of neutrophil elastase in pulmonary emphysema, neutrophil elastase-deficient mice and wild-type littermate controls were exposed to long-term cigarette smoke. Compared to wild-type littermates, mice that were deficient in neutrophil elastase were significantly protected (59%) from the development of emphysema. Previously, we demonstrated complete protection from emphysema in the absence of macrophage elastase. Further analysis revealed several interactions between these two elastases. Each elastase inactivated the endogenous inhibitor of the other, with neutrophil elastase degrading tissue inhibitor of metalloproteinase-1, and macrophage elastase degrading alpha-1-antitrypsin. Cigarette smoke-induced recruitment of both neutrophils and monocytes was impaired in the absence of neutrophil elastase. Moreover, there was less macrophage elastase activity secondary to decreased macrophage accumulation in neutrophil elastase-deficient mice. This study demonstrates a direct role for neutrophil elastase in emphysema and highlights the interdependence of the proteinases and inflammatory cells that mediate lung destruction in response to cigarette smoke.


Subject(s)
Leukocyte Elastase/metabolism , Nicotiana , Pulmonary Emphysema/enzymology , Pulmonary Emphysema/etiology , Smoke/adverse effects , Animals , Macrophages/metabolism , Matrix Metalloproteinase 12 , Metalloendopeptidases/metabolism , Mice , Mice, Knockout , Monocytes/pathology , Neutrophil Infiltration , Pancreatic Elastase/metabolism , Pulmonary Emphysema/pathology , Pulmonary Emphysema/physiopathology , Time Factors
16.
J Clin Invest ; 109(3): 363-71, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11827996

ABSTRACT

Leukocyte recruitment in inflammation is critical for host defense, but excessive accumulation of inflammatory cells can lead to tissue damage. Neutrophil-derived serine proteases (cathepsin G [CG], neutrophil elastase [NE], and proteinase 3 [PR3]) are expressed specifically in mature neutrophils and are thought to play an important role in inflammation. To investigate the role of these proteases in inflammation, we generated a mouse deficient in dipeptidyl peptidase I (DPPI) and established that DPPI is required for the full activation of CG, NE, and PR3. Although DPPI(-/-) mice have normal in vitro neutrophil chemotaxis and in vivo neutrophil accumulation during sterile peritonitis, they are protected against acute arthritis induced by passive transfer of monoclonal antibodies against type II collagen. Specifically, there is no accumulation of neutrophils in the joints of DPPI(-/-) mice. This protective effect correlates with the inactivation of neutrophil-derived serine proteases, since NE(-/-) x CG(-/-) mice are equally resistant to arthritis induction by anti-collagen antibodies. In addition, protease-deficient mice have decreased response to zymosan- and immune complex-mediated inflammation in the subcutaneous air pouch. This defect is accompanied by a decrease in local production of TNF-alpha and IL-1 beta. These results implicate DPPI and polymorphonuclear neutrophil-derived serine proteases in the regulation of cytokine production at sites of inflammation.


Subject(s)
Arthritis, Experimental/enzymology , Arthritis, Experimental/etiology , Cathepsin C/metabolism , Neutrophils/enzymology , Serine Endopeptidases/metabolism , Animals , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Cathepsin C/deficiency , Cathepsin C/genetics , Cathepsin G , Cathepsins/deficiency , Cathepsins/genetics , Cathepsins/metabolism , Cytokines/biosynthesis , Enzyme Activation , Hematopoiesis , Interleukin-1/biosynthesis , Leukocyte Elastase/deficiency , Leukocyte Elastase/genetics , Leukocyte Elastase/metabolism , Mice , Mice, Knockout , Myeloblastin , Neutrophils/pathology , Tumor Necrosis Factor-alpha/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL