@article{white_martin_stern_laxman_marroquin_2010, title={Expression of IL-4/IL-13 receptors in differentiating human airway epithelial cells}, volume={299}, ISSN={["1522-1504"]}, DOI={10.1152/ajplung.00422.2009}, abstractNote={IL-4 and IL-13 elicit several important responses in airway epithelium including chemokine secretion and mucous secretion that may contribute to airway inflammation, cell migration, and differentiation. These cytokines have overlapping but not identical effector profiles likely due to shared subunits in their receptor complexes. These receptors are variably described in epithelial cells, and the relative expression, localization, and function of these receptors in differentiated and repairing epithelial cells are not clear. We examined IL-4/IL-13 receptor expression and localization in primary airway epithelial cells collected from normal human lungs and grown under conditions yielding both undifferentiated and differentiated cells inclusive of basal, goblet, and ciliated cell phenotypes. Gene expression of the IL-4Rα, IL-2Rγc, IL-13Rα1, and IL-13Rα2 receptor subunits increased with differentiation, but different patterns of localization and protein abundance were seen for each subunit based on both differentiation and the cell subtypes present. Increased expression of receptor subunits observed in more differentiated cells was associated with more substantial functional responses to IL-4 stimulation including increased eotaxin-3 expression and accelerated migration after injury. We demonstrate substantial differences in IL-4/IL-13 receptor subunit expression and responsiveness to IL-4 based on the extent of airway epithelial cell differentiation and suggest that these differences may have functional consequences in airway inflammation.}, number={5}, journal={AMERICAN JOURNAL OF PHYSIOLOGY-LUNG CELLULAR AND MOLECULAR PHYSIOLOGY}, author={White, Steven R. and Martin, Linda D. and Stern, Randi and Laxman, Bharathi and Marroquin, Bertha A.}, year={2010}, month={Nov}, pages={L681–L693} } @article{manzo_slade_richards_mcgee_martin_dye_2010, title={Susceptibility of Inflamed Alveolar and Airway Epithelial Cells to Injury Induced by Diesel Exhaust Particles of Varying Organic Carbon Content}, volume={73}, ISSN={["1087-2620"]}, DOI={10.1080/15287390903566625}, abstractNote={Exposure to traffic-related ambient air pollution, such as diesel exhaust particles (DEP), is associated with adverse health outcomes, especially in individuals with preexisting inflammatory respiratory diseases. Using an analogous novel in vitro system to model both the healthy and inflamed lung, the susceptibility of epithelial cells exposed to DEP of varying organic carbon content was studied. Murine LA-4 alveolar type II-like epithelial cells, as well as primary murine tracheal epithelial cells (MTE), were treated with exogenous cytokines (tumor necrosis factor [TNF] α + interleukin [IL]-1 β + interferon [IFN] γ) to model a mild inflammatory state. Epithelial cells were subsequently exposed to DEP of varying organic carbon content, and the resultant cytotoxic, cytoprotective, or antioxidant cell responses were inferred by changes in lactate dehydrogenase (LDH) release, heme oxygenase-1 (HO-1) expression, or glutathione levels, respectively. Data showed that exposure of healthy LA-4 cells to organic carbon-rich DEP (25 μg/cm2; 24 h) induced adaptive cytoprotective/antioxidant responses with no apparent cell injury. In contrast, exposure of inflamed LA-4 cells resulted in oxidative stress culminating in significant cytotoxicity. Exposure of healthy MTE cells to organic carbon-rich DEP (20 μg/cm2; 24 h) was seemingly without effect, whereas exposure of inflamed MTE cells resulted in increased epithelial solute permeability. Thus, surface lung epithelial cells stressed by a state of inflammation and then exposed to organic carbon-rich DEP appear unable to respond to the additional oxidative stress, resulting in epithelial barrier dysfunction and injury. Adverse health outcomes associated with exposure to traffic-related air pollutants, like DEP, in patients with preexisting inflammatory respiratory diseases may be due, in part, to similar mechanisms.}, number={8}, journal={JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH-PART A-CURRENT ISSUES}, author={Manzo, Nicholas D. and Slade, Ralph and Richards, Judy H. and McGee, John K. and Martin, Linda D. and Dye, Janice A.}, year={2010}, pages={565–580} } @article{white_martin_abe_marroquin_stern_fu_2009, title={Insulin receptor substrate-1/2 mediates IL-4-induced migration of human airway epithelial cells}, volume={297}, ISSN={["1040-0605"]}, DOI={10.1152/ajplung.90453.2008}, abstractNote={Migration of airway epithelial cells (AEC) is an integral component of airway mucosal repair after injury. The inflammatory cytokine IL-4, abundant in chronic inflammatory airways diseases such as asthma, stimulates overproduction of mucins and secretion of chemokines from AEC; these actions enhance persistent airway inflammation. The effect of IL-4 on AEC migration and repair after injury, however, is not known. We examined migration in primary human AEC differentiated in air-liquid interface culture for 3 wk. Wounds were created by mechanical abrasion and followed to closure using digital microscopy. Concurrent treatment with IL-4 up to 10 ng/ml accelerated migration significantly in fully differentiated AEC. As expected, IL-4 treatment induced phosphorylation of the IL-4 receptor-associated protein STAT (signal transducer and activator of transcription)6, a transcription factor known to mediate several IL-4-induced AEC responses. Expressing a dominant negative STAT6 cDNA delivered by lentivirus infection, however, failed to block IL-4-stimulated migration. In contrast, decreasing expression of either insulin receptor substrate (IRS)-1 or IRS-2 using a silencing hairpin RNA blocked IL-4-stimulated AEC migration completely. These data demonstrate that IL-4 can accelerate migration of differentiated AEC after injury. This reparative response does not require STAT6 activation, but rather requires IRS-1 and/or IRS-2.}, number={1}, journal={AMERICAN JOURNAL OF PHYSIOLOGY-LUNG CELLULAR AND MOLECULAR PHYSIOLOGY}, author={White, Steven R. and Martin, Linda D. and Abe, Mark K. and Marroquin, Bertha A. and Stern, Randi and Fu, Xiaoying}, year={2009}, month={Jul}, pages={L164–L173} } @article{booth_sandifer_martin_martin_2007, title={IL-13-induced proliferation of airway epithelial cells: mediation by intracellular growth factor mobilization and ADAM17}, volume={8}, journal={Respiratory Research}, author={Booth, B. W. and Sandifer, T. and Martin, E. L. and Martin, L. D.}, year={2007} } @misc{li_martin_adler_2007, title={Method and compositions for altering mucus secretion}, volume={7,265,088}, number={2007 Sept. 4}, publisher={Washington, DC: U.S. Patent and Trademark Office}, author={Li, Y.-H. and Martin, L. D. and Adler, K. B.}, year={2007} } @article{chorley_crews_li_adler_minnicozzi_martin_2006, title={Differential Muc2 and Muc5ac secretion by stimulated guinea pig tracheal epithelial cells in vitro}, volume={7}, journal={Respiratory Research}, author={Chorley, B. N. and Crews, A. L. and Li, Y. H. and Adler, K. B. and Minnicozzi, M. and Martin, L. D.}, year={2006}, pages={35} } @misc{takashi_parikh_adler_martin_y._2006, title={Methods for regulating inflammatory mediators and peptides useful therein}, volume={7,544,772}, number={2006 Sep. 28}, publisher={Washington, DC: U.S. Patent and Trademark Office}, author={Takashi, S. and Parikh, I. and Adler, K. B. and Martin, L. D. and Y., Li}, year={2006} } @article{mexas_hess_hawkins_martin_2006, title={Pulmonary lesions in cats with diabetes mellitus}, volume={20}, ISSN={["1939-1676"]}, DOI={10.1892/0891-6640(2006)20[47:PLICWD]2.0.CO;2}, abstractNote={Diabetes mellitus (DM) is a common endocrinopathy of cats and humans. Although few studies have examined the effects of DM on the pulmonary system, changes in pulmonary function and immunology in humans with type I and II diabetes, and pulmonary lesions in a murine diabetic model have been documented. Our objective was to determine whether pulmonary lesions occurred in cats with DM. Medical records and necropsy evaluations of 42 cats with DM were compared with those of 45 age-matched, nondiabetic cats for the presence of clinical evidence of respiratory disease and pulmonary histopathological findings at the time of necropsy. No statistical difference was noted in the presence of clinical evidence of respiratory disease between cats with diabetes and control cats. Nevertheless, there was a significant association between the presence of abnormal pulmonary histopathology and DM (P= .018, odds ratio = 3 inclusive of all cats; P= .005, odds ratio = 5 when non-DM cats with overt clinical evidence of respiratory disease were excluded). Pulmonary abnormalities detected by histopathological examination in cats with diabetes included congestion and edema, histiocytosis, pneumonia, smooth muscle hypertrophy, fibrosis, mineralization, neoplasia, and type II pneumocyte hyperplasia. The observed association between DM and pulmonary lesions in cats, independent of clinical evidence of respiratory disease, emphasizes the need for careful assessment of the respiratory tract in sick cats with diabetes.}, number={1}, journal={JOURNAL OF VETERINARY INTERNAL MEDICINE}, author={Mexas, AM and Hess, RS and Hawkins, EC and Martin, LD}, year={2006}, pages={47–51} } @misc{martin_adler_macchione_akley_mckane_2005, title={Culture system for mouse tracheal epithelial cells}, volume={6,933,149}, number={2005 Aug. 23}, publisher={Washington, DC: U.S. Patent and Trademark Office}, author={Martin, L. D. and Adler, K. B. and Macchione, M. and Akley, N. J. and McKane, S. A.}, year={2005} } @article{fischer_cuellar_byrd_rice_bonner_martin_voynow_2005, title={ErbB2 activity is required for airway epithelial repair following neutrophil elastase exposure}, volume={19}, number={7}, journal={FASEB Journal}, author={Fischer, B. M. and Cuellar, J. G. and Byrd, A. S. and Rice, A. B. and Bonner, J. C. and Martin, L. D. and Voynow, J. A.}, year={2005} } @article{park_he_martin_li_chorley_adler_2005, title={Human neutrophil elastase induces hypersecretion of mucin from well-differentiated human bronchial epithelial cells in vitro via a protein kinase C delta-mediated mechanism}, volume={167}, ISSN={["1525-2191"]}, DOI={10.1016/S0002-9440(10)62040-8}, abstractNote={The presence of mucus obstruction and neutrophil-predominant inflammation in several lung disorders, such as cystic fibrosis, suggests a relationship between neutrophils and excess mucus production. Mechanisms of human neutrophil elastase (HNE)-induced mucin secretion by well-differentiated normal human bronchial epithelial (NHBE) cells maintained in air/liquid interface culture were investigated. HNE increased mucin secretion in a concentration-dependent manner, with maximal stimulation (more than twofold) occurring within a short (15 minutes) time period. Mucins MUC5AC and MUC5B, but not MUC2, were released in response to HNE. Stimulation of mucin secretion required partial elastase enzymatic activity and did not appear to involve a soluble product released by the cells. HNE-stimulated secretion involved activation of protein kinase C (PKC), as HNE exposure rapidly provoked PKC enzymatic activity that was attenuated by the general PKC inhibitors calphostin C and bisindoylmaleimide I. Of the different isoforms, PKCα, δ, ζ, λ, ι, and ε were constitutively expressed in NHBE cells while PKCβ, η, and μ were PMA-inducible. PKCδ was the only isoform to translocate from cytoplasm to membrane in response to HNE. Inhibition of PKCδ attenuated HNE-mediated mucin secretion. The results suggest HNE stimulation of mucin release by human airway epithelial cells involves intracellular activation of PKC, specifically the δ isoform. The presence of mucus obstruction and neutrophil-predominant inflammation in several lung disorders, such as cystic fibrosis, suggests a relationship between neutrophils and excess mucus production. Mechanisms of human neutrophil elastase (HNE)-induced mucin secretion by well-differentiated normal human bronchial epithelial (NHBE) cells maintained in air/liquid interface culture were investigated. HNE increased mucin secretion in a concentration-dependent manner, with maximal stimulation (more than twofold) occurring within a short (15 minutes) time period. Mucins MUC5AC and MUC5B, but not MUC2, were released in response to HNE. Stimulation of mucin secretion required partial elastase enzymatic activity and did not appear to involve a soluble product released by the cells. HNE-stimulated secretion involved activation of protein kinase C (PKC), as HNE exposure rapidly provoked PKC enzymatic activity that was attenuated by the general PKC inhibitors calphostin C and bisindoylmaleimide I. Of the different isoforms, PKCα, δ, ζ, λ, ι, and ε were constitutively expressed in NHBE cells while PKCβ, η, and μ were PMA-inducible. PKCδ was the only isoform to translocate from cytoplasm to membrane in response to HNE. Inhibition of PKCδ attenuated HNE-mediated mucin secretion. The results suggest HNE stimulation of mucin release by human airway epithelial cells involves intracellular activation of PKC, specifically the δ isoform. Neutrophils are involved in a variety of inflammatory lung disorders including chronic bronchitis, bronchiectasis, cystic fibrosis, and probably asthma. In these diseases, the pathological findings of mucus obstruction and neutrophil-predominant inflammation in airways1Fahy JV Kim KW Liu J Boushey HA Prominent neutrophilic inflammation in sputum from subjects with asthma exacerbation.J Allergy Clin Immunol. 1995; 95: 843-852Abstract Full Text Full Text PDF PubMed Scopus (568) Google Scholar, 2Stockley RA Role of inflammation in respiratory tract infections.Am J Med. 1995; 99: 8S-13SAbstract Full Text PDF PubMed Scopus (47) Google Scholar, 3Welsh MD Adair BM Foster JC Effect of BVD virus infection on alveolar macrophage functions.Vet Immunol Immunopathol. 1995; 46: 195-210Crossref PubMed Scopus (54) Google Scholar, 4Mohapatra NK Cheng PW Parker JC Paradiso AM Yankaskas JR Boucher RC Boat TF Alteration of sulfation of glycoconjugates, but not sulfate transport and intracellular inorganic sulfate content in cystic fibrosis airway epithelial cells.Pediatr Res. 1995; 38: 42-48Crossref PubMed Scopus (28) Google Scholar, 5Fahy JV Schuster A Ueki I Boushey HA Nadel JA Mucus hypersecretion in bronchiectasis. The role of neutrophil proteases.Am Rev Respir Dis. 1992; 146: 1430-1433Crossref PubMed Scopus (109) Google Scholar, 6Stockley RA Hill SL Morrison HM Starkie CM Elastolytic activity of sputum and its relation to purulence and to lung function in patients with bronchiectasis.Thorax. 1984; 39: 408-413Crossref PubMed Scopus (93) Google Scholar suggest a relationship between neutrophil recruitment/infiltration and excess mucus production and secretion. Neutrophils store three proteases that have been implicated in airway mucin secretion: elastase,7Breuer R Christensen TG Lucey EC Stone PJ Snider GL An ultrastructural morphometric analysis of elastase-treated hamster bronchi shows discharge followed by progressive accumulation of secretory granules.Am Rev Respir Dis. 1987; 136: 698-703Crossref PubMed Scopus (46) Google Scholar, 8Nadel JA Protease actions on airway secretions. Relevance to cystic fibrosis.Ann NY Acad Sci. 1991; 624: 286-296Crossref PubMed Scopus (25) Google Scholar, 9Kim KC Wasano K Niles RM Schuster JE Stone PJ Brody JS Human neutrophil elastase releases cell surface mucins from primary cultures of hamster tracheal epithelial cells.Proc Natl Acad Sci USA. 1987; 84: 9304-9308Crossref PubMed Scopus (124) Google Scholar cathepsin G,10Sommerhoff CP Nadel JA Basbaum CB Caughey GH Neutrophil elastase and cathepsin G stimulate secretion from cultured bovine airway gland serous cells.J Clin Invest. 1990; 85: 682-689Crossref PubMed Scopus (285) Google Scholar and proteinase-3.11Rao NV Marshall BC Gray BH Hoidal JR Interaction of secretory leukocyte protease inhibitor with proteinase-3.Am J Respir Cell Mol Biol. 1993; 8: 612-616Crossref PubMed Scopus (60) Google Scholar, 12Renesto P Halbwachs-Mecarelli L Nusbaum P Lesavre P Chignard M Proteinase 3. A neutrophil proteinase with activity on platelets.J Immunol. 1994; 152: 4612-4617PubMed Google Scholar Of these, human neutrophil elastase (HNE), a major component of primary or azurophilic granules,13Bainton DF Ullyot JL Farquhar MG The development of neutrophilic polymorphonuclear leukocytes in human bone marrow.J Exp Med. 1971; 134: 907-934Crossref PubMed Scopus (565) Google Scholar is the most widely studied with regard to enhanced mucus secretion. Levels of HNE are elevated in airways of patients with chronic bronchitis and cystic fibrosis,14Fick Jr, RB Naegel GP Squier SU Wood RE Gee JB Reynolds HY Proteins of the cystic fibrosis respiratory tract. Fragmented immunoglobulin G opsonic antibody causing defective opsonophagocytosis.J Clin Invest. 1984; 74: 236-248Crossref PubMed Scopus (153) Google Scholar and levels in patients' sputum may exceed 100 μg/ml (3.3 × 10−6 mol/L).15Doring G Goldstein W Botzenhart K Kharazmi A Schiotz PO Hoiby N Dasgupta M Elastase from polymorphonuclear leucocytes: a regulatory enzyme in immune complex disease.Clin Exp Immunol. 1986; 64: 597-605PubMed Google Scholar, 16Goldstein W Doring G Lysosomal enzymes from polymorphonuclear leukocytes and proteinase inhibitors in patients with cystic fibrosis.Am Rev Respir Dis. 1986; 134: 49-56PubMed Google Scholar, 17Suter S Schaad UB Tegner H Ohlsson K Desgrandchamps D Waldvogel FA Levels of free granulocyte elastase in bronchial secretions from patients with cystic fibrosis: effect of antimicrobial treatment against Pseudomonas aeruginosa.J Infect Dis. 1986; 153: 902-909Crossref PubMed Scopus (104) Google Scholar Purified HNE has been shown to provoke secretion of mucin by isolated airway epithelial cells and glands from several species.7Breuer R Christensen TG Lucey EC Stone PJ Snider GL An ultrastructural morphometric analysis of elastase-treated hamster bronchi shows discharge followed by progressive accumulation of secretory granules.Am Rev Respir Dis. 1987; 136: 698-703Crossref PubMed Scopus (46) Google Scholar, 8Nadel JA Protease actions on airway secretions. Relevance to cystic fibrosis.Ann NY Acad Sci. 1991; 624: 286-296Crossref PubMed Scopus (25) Google Scholar, 10Sommerhoff CP Nadel JA Basbaum CB Caughey GH Neutrophil elastase and cathepsin G stimulate secretion from cultured bovine airway gland serous cells.J Clin Invest. 1990; 85: 682-689Crossref PubMed Scopus (285) Google Scholar, 18Kim KC Nassiri J Brody JS Mechanisms of airway goblet cell mucin release: studies with cultured tracheal surface epithelial cells.Am J Respir Cell Mol Biol. 1989; 1: 137-143Crossref PubMed Scopus (46) Google Scholar Although there have been suggestions that interactions between HNE and epithelial cell surfaces may be involved in the response,9Kim KC Wasano K Niles RM Schuster JE Stone PJ Brody JS Human neutrophil elastase releases cell surface mucins from primary cultures of hamster tracheal epithelial cells.Proc Natl Acad Sci USA. 1987; 84: 9304-9308Crossref PubMed Scopus (124) Google Scholar, 19Takeyama K Agusti C Ueki I Lausier J Cardell LO Nadel JA Neutrophil-dependent goblet cell degranulation: role of membrane-bound elastase and adhesion molecules.Am J Physiol. 1998; 275: L294-L302PubMed Google Scholar intracellular mechanisms and signaling pathways associated with HNE-induced mucin hypersecretion have not been elucidated. In this study, well-differentiated primary normal human tracheobronchial epithelial (NHBE) cells maintained in vitro in air/liquid interface were exposed to HNE, and the secretory response assessed. Elastase proved to be a potent mucin secretagogue for NHBE cells, eliciting a robust (greater than twofold) increase in mucin secretion within 15 minutes. The mucin gene products released included those of MUC5AC and MUC5B, but not of MUC2. The mechanism appeared to involve activation of protein kinase C (PKC), as HNE exposure rapidly provoked phosphorylation of MARCKS (myristoylated alanine-rich C kinase substrate) protein, a cellular substrate of PKC, and the mucin secretory response to HNE was attenuated by two different PKC inhibitors. Additional studies provided compelling evidence that PKCδ is the specific PKC isoform involved in the secretory pathway. All chemicals were of analytical grade or higher. NHBE cells, bronchial epithelial basal medium, and supplements for air/liquid interface cell cultures were purchased from Cambrex (San Diego, CA). Endotoxin-free HNE purified from human sputum was purchased from Elastin Products Company (EPC, Owensville, MO). Cytotoxicity was evaluated with CytoTox 96 nonradioactive cytotoxicity assay kits obtained from Promega Corp. (Madison, WI). A specific HNE substrate, MeO-SUC-AL-AL-PRO-VAL-PNA, and an HNE inhibitor, chloromethyl ketone-modified tetrapeptide (CMK), also were purchased from EPC and the HNE inhibitor elastatinal was obtained from Calbiochem (La Jolla, CA). 17Q2 pan mucin antibody was purchased from Babco (Richmond, CA) and anti-MUC5AC (45M1) was purchased from Neomarkers (Fremont, CA). A monoclonal antibody (11C1) against human MUC5B was generously provided by Dr. Reen Wu, University of California at Davis, Davis, CA. The epitope for this antibody, which was generated from the secreted mucin of well-differentiated airway epithelial cells, is not known, but by immunohistochemical staining and Western blot analysis, it appears to recognize the MUC5B peptide. A monoclonal antibody that cross reacts with human MUC2, raised against the guinea pig 522-bp gene sequence analogous to the human D4 domain located in the carboxy-terminal region of the Muc2 gene sequence established previously in our laboratory, was used to detect MUC2 mucins.20Li Y Martin LD Minnicozzi M Greenfeder S Fine J Pettersen CA Chorley B Adler KB Enhanced expression of mucin genes in a guinea pig model of allergic asthma.Am J Respir Cell Mol Biol. 2001; 25: 644-651Crossref PubMed Scopus (32) Google Scholar An ImmunoPure (G) IgG purification kit used for purification of antibodies for enzyme-linked immunosorbent assay (ELISA) was from Pierce (Rockford, IL). For Western blot analysis of PKC isoforms expressed in NHBE cells, a PKC sampler kit and E-cadherin antibody were obtained from BD Biosciences (San Jose, CA). Goat anti-PKCζ and mouse anti-α-tubulin were purchased from Santa Cruz Biotechnology (Santa Cruz, CA). Antibodies against phosphorylated (ser) PKC substrate and phosphorylated MARCKS were from Cell Signaling Technology (Beverly, MA). Horseradish peroxidase-conjugated goat anti-mouse IgG and donkey anti-goat IgG also were purchased from Santa Cruz Biotechnology. Horseradish peroxidase-conjugated goat anti-rabbit IgG was purchased from Upstate Biotechnology (Lake Placid, NY). Enhanced chemiluminescence development kits and Hyperfilm were from Amersham Pharmacia Biotech (Piscataway, NJ). All PKC-related inhibitors (ie, calphostin C, bisindoylmaleimide, PKC epsilon and zeta inhibitor peptides, rottlerin) were purchased from Calbiochem. A PepTag assay for nonradioactive detection of PKC activity was purchased from Promega. Other chemical reagents were purchased from Sigma-Aldrich (St. Louis, MO). Transwell-Clear culture inserts and high-binding 96-well assay plates were purchased from Corning Inc. (Corning, NY). Primary cultures of NHBE cells were established using an air/liquid interface cell culture system described previously.21Li Y Martin LD Spizz G Adler KB MARCKS protein is a key molecule regulating mucin secretion by human airway epithelial cells in vitro.J Biol Chem. 2001; 276: 40982-40990Crossref PubMed Scopus (155) Google Scholar Briefly, NHBE cells were expanded once and cells collected and frozen in liquid nitrogen (referred to as passage-2 cells). Air/liquid interface cultures of NHBE cells were established on Transwell-Clear culture inserts thin-coated with rat-tail type I collagen. The basic medium used for NHBE cells was a 1:1 mixture of bronchial epithelial basal medium and high glucose (4.5 g/L) Dulbecco's modified Eagle's medium. The complete medium was composed of basic medium containing a final concentration of 0.5 ng/ml human recombinant epidermal growth factor, 0.5 μg/ml hydrocortisone, 5 μg/ml insulin, 10 μg/ml transferrin, 0.5 μg/ml epinephrine, 6.5 ng/ml triiodothyronine, 50 μg/ml gentamicin, and 50 ng/ml amphotericin-B. In addition, the media contained 0.13 mg/ml bovine pituitary extract made according to the protocol of Bertolero and colleagues,22Bertolero F Kaighn ME Gonda MA Saffiotti U Mouse epidermal keratinocytes. Clonal proliferation and response to hormones and growth factors in serum-free medium.Exp Cell Res. 1984; 155: 64-80Crossref PubMed Scopus (61) Google Scholar 5 × 10−8 mol/L all-trans retinoic acid, 1.5 μg/ml bovine serum albumin, and 20 U/ml nystatin. Frozen NHBE cells were recovered and seeded at a density of ∼2 × 104 cells/cm2 onto the apical surface of the inserts. Media were changed the next day, then every other day until the cells reached ∼90% confluence. At this point, the air/liquid interface was established by removing the apical media, whereas basolateral media were changed daily for up to 21 days. A mucin phenotype was observed at ∼14 days in culture (∼7 days in air-liquid interface culture) and cilia were apparent by 18 days in culture. Mucin secretion reached maximal levels at ∼18 days in culture, so cells cultured for ∼18 to 21 days were used for the experiments described below. HNE stock was made as 10 mg/ml (339 μmol/L) in a 1:1 mixture of glycerol and 0.02 mol/L NaOAc, pH 5.0. The stock was diluted into the culture medium to the final concentration indicated. In all studies, the above solvent appropriately diluted was used as a negative control. NHBE cells were exposed to HNE from both apical and basolateral sides for 15 minutes (unless otherwise indicated). At the end of each treatment, apical medium containing the secreted mucin was collected and quantified. Briefly, 0.25 ml of media containing secreted mucin was collected, 0.5 ml of 1 mmol/L dithiothreitol in phosphate-buffered saline (PBS) was added into each well, and the plates were gently agitated and allowed to stand for 3 minutes before the dithiothreitol/PBS plus mucin was collected in the same tube. Finally, 0.5 ml of 10 μmol/L CMK in PBS was added and collected the same way. Approximately 1.25 ml of the collected mucin mixture with dithiothreitol and CMK was centrifuged at 8000 rpm for 5 minutes to remove cell debris, and then collected in a fresh tube. Phenylmethyl sulfonyl fluoride was added to a final concentration of 1 mmol/L. Baseline and treatment mucin secretions were collected from each culture plate. Baseline mucin secretion was collected to normalize variations from well to well, and to control for possible release of mucin in response to the stress of media change or washing. After the baseline mucin secretion sample was collected, the cells were rested overnight and exposed to test agents the next day for indicated periods of time. Mucin samples were quantified using specific ELISA methods. Firstly, total mucin was quantified by a double-sandwich ELISA using a pan-mucin antibody, 17Q2, that cross reacts with a carbohydrate epitope on human mucins, as described previously.21Li Y Martin LD Spizz G Adler KB MARCKS protein is a key molecule regulating mucin secretion by human airway epithelial cells in vitro.J Biol Chem. 2001; 276: 40982-40990Crossref PubMed Scopus (155) Google Scholar Additional studies were performed using ELISAs for secreted protein products of the mucin genes MUC5AC, MUC5B, and MUC2 to determine which mucin gene products were being released on exposure to HNE. MUC5AC was measured via ELISA as described by Takeyama and colleagues23Takeyama K Dabbagh K Lee HM Agusti C Lausier JA Ueki IF Grattan KM Nadel JA Epidermal growth factor system regulates mucin production in airways.Proc Natl Acad Sci USA. 1999; 96: 3081-3086Crossref PubMed Scopus (522) Google Scholar using the 45M1 antibody. MUC5B protein was assayed via a standard double-sandwich ELISA method using the 11C1 monoclonal antibody against MUC5B provided by Dr. Reen Wu, University of California, Davis, Davis, CA, as described previously.24Groneberg DA Eynott PR Oates T Lim S Wu R Carlstedt L Nicholson AG Chung KF Expression of MUC5AC and MUC5B mucins in normal and cystic fibrosis lung.Respir Med. 2002; 96: 81-86Abstract Full Text PDF PubMed Scopus (154) Google Scholar, 25Crowther JR ELISA. Theory and practice.Methods Mol Biol. 1995; 42: 1-218PubMed Google Scholar The MUC2 gene product was quantified by modification of an ELISA as described previously.20Li Y Martin LD Minnicozzi M Greenfeder S Fine J Pettersen CA Chorley B Adler KB Enhanced expression of mucin genes in a guinea pig model of allergic asthma.Am J Respir Cell Mol Biol. 2001; 25: 644-651Crossref PubMed Scopus (32) Google Scholar HNE activity assays were performed following the manufacturer's protocol (EPC). HNE substrate was prepared in substrate buffer (Tris-NaCl buffer: 0.1 mol/L Tris, pH 7.5, containing 0.5 mol/L NaCl and 0.01% Na3N). Briefly, 3 ml of substrate solution at 25°C was added to test tubes, 1.0 μg of HNE then was added, and the developed color was read immediately and continuously thereafter at 1 minute intervals. Elastase activity was reflected by the rate increase in absorbance in time units (minutes). Color development was read at 410 nm on a spectrophotometer UV160U (Shimadzu, Kyoto, Japan). The specific activity of HNE was expressed as U/mg, and results expressed as percentage of activity of native HNE for each treatment. Effects of enzymatic inhibition of HNE were investigated using three different elastase inhibitors: 1) elastatinal, a natural HNE inhibitor produced by Actinomycetes;26Umezawa H Structures and activities of protease inhibitors of microbial origin.Methods Enzymol. 1976; 45: 678-695Crossref PubMed Scopus (303) Google Scholar 2) CMK, a synthetic tetrapeptide;27Rees DD Brain JD Wohl ME Humes JL Mumford RA Inhibition of neutrophil elastase in CF sputum by L-658,758.J Pharmacol Exp Ther. 1997; 283: 1201-1206PubMed Google Scholar and 3) α1-antitrypsin (α1-AT), a physiological HNE inhibitor.28Gadek JE Fells GA Zimmerman RL Rennard SI Crystal RG Antielastases of the human alveolar structures. Implications for the protease-antiprotease theory of emphysema.J Clin Invest. 1981; 68: 889-898Crossref PubMed Scopus (311) Google Scholar The inhibitors were added directly to HNE, incubated for 15 minutes at 37°C, and then added directly to the cells for another 15 minutes. At the end of this exposure, secreted mucin was collected and quantified as described above. To determine whether HNE enzymatic activity was directly required for stimulated mucin secretion, or if a secondary product(s) released by NHBE cells after exposure to HNE could be involved in the secretory response, NHBE cells were exposed to HNE (or vehicle) for 5 minutes. After exposure, the conditioned medium was collected and treated with 5 μmol/L of the HNE enzymatic inhibitor, α1-AT, for 15 minutes, at which time this α1-AT-treated medium was added to a new set of NHBE cells and effects on mucin secretion quantified as described above. The PKC inhibitors, bisindolylmaleimide I (10, 100, 1000 nmol/L)29Martiny-Baron G Kazanietz MG Mischak H Blumberg PM Kochs G Hug H Marme D Schachtele C Selective inhibition of protein kinase C isozymes by the indolocarbazole Go 6976.J Biol Chem. 1993; 268: 9194-9197Abstract Full Text PDF PubMed Google Scholar or calphostin C (5, 50, 500 nmol/L)30Takahashi I Saitoh Y Yoshida M Sano H Nakano H Morimoto M Tamaoki T UCN-01 and UCN-02, new selective inhibitors of protein kinase C. II. Purification, physico-chemical properties, structural determination and biological activities.J Antibiot (Tokyo). 1989; 42: 571-576Crossref PubMed Scopus (168) Google Scholar were used to determine PKC involvement in HNE-induced mucin secretion. NHBE cells were preincubated with these agents (or vehicle control) for 15 minutes, then HNE was added for another 15 minutes before mucin secretion was quantified as described above. PKC activity in NHBE cells after exposure to HNE was assessed using a PepTag assay for nonradioactive detection of PKC (following the manufacturer's protocol). Briefly, 10 μg of protein extracted from each treatment of NHBE cells was added into the PKC reaction buffer (20 mmol/L HEPES, pH 7.4, 1.3 mmol/L CaCl2, 1 mmol/L dithiothreitol, 10 mmol/L MgCl2, 1 mmol/L ATP) containing 1 mg/ml phosphatidylserine and PepTag C1 PKC substrate peptide (P-L-S-R-T-L-S-V-A-A-K) conjugated with fluorescent dye, and incubated for 30 minutes at 30°C. The reaction was stopped by boiling at 100°C for 10 minutes. Reaction mixtures were separated on 0.8% agarose gels and proteins quantified by Labworks image acquisition and analysis software (UVP Bioimaging System, Upland, CA). Phosphorylation of MARCKS was detected by Western blot using an antibody against phophospecific-MARCKS. After treatments, NHBE cells were washed with ice-cold PBS twice and then scraped into lysis buffer (50 mmol/L Tris, pH 7.5, 1 mmol/L ethylenediamine tetraacetic acid, 100 mmol/L NaCl, 1 mmol/L phenylmethyl sulfonyl fluoride) using a rubber policemen. The collected cells were lysed by sonication. For separation of cytosolic and membrane fractions, the lysates were spun at 400,000 × g in a Sorvall Discovery 100S ultracentrifuge (Sorvall, Inc. Newtown, CT) for 1 hour. The supernatant was reserved as the cytosolic sample. The pellet was resuspended in the same lysis buffer containing 0.05% Triton-100, dissolved by sonication, and incubated on ice for 30 minutes. After incubation, the same ultracentrifugation as described above was performed on the pellet mixture, and the supernatant separated from the pellet mixture was reserved as the membrane fraction. For preparation of whole cell crude lysates, the disrupted cellular mixture was centrifuged at 15,000 rpm in an Eppendorf 5417R centrifuge (Eppendorf Corp., Hamburg, Germany) for 1 hour at 4°C. The supernatant was collected as the whole crude NHBE cell lysate. The protein concentration of cell lysate samples was quantified by a Bradford assay (Bio-Rad Laboratories, Hercules, CA). Each sample was boiled in 2× sodium dodecyl sulfate-polyacrylamide gel electrophoresis sample buffer for 10 minutes, loaded on 10% sodium dodecyl sulfate-polyacrylamide gel electrophoresis gels, and transferred to a polyvinylidene difluoride membrane (Micron Separation Inc., Westborough, MA). After blocking with 5% skim milk, the antigen was captured by the specific PKC antibody and further amplified by binding to horseradish peroxidase-conjugated anti-mouse or anti-rabbit antibodies. Anti-α-tubulin and E-cadherin antibodies were used for cytosolic and membrane controls, respectively, for each sample. Final development was accomplished by the enhanced chemiluminescence method. The amount of each PKC isoform was analyzed by Labworks image acquisition and analysis software. Because the studies above indicated that PKCδ was the only isoform to translocate to membranes in response to HNE, additional studies were performed with rottlerin, an inhibitor of PKCδ and θ.31Gschwendt M Muller HJ Kielbassa K Zang R Kittstein W Rincke G Marks F Rottlerin, a novel protein kinase inhibitor.Biochem Biophys Res Commun. 1994; 199: 93-98Crossref PubMed Scopus (767) Google Scholar (Because PKCθ was not expressed in NHBE cells under basal or stimulated conditions, rottlerin is referred to below as a specific inhibitor of PKCδ). Rottlerin has the following potency against PKC isoforms: PKC δ (IC50 = 3 to 6 μmol/L); PKCθ (IC50 = 50 μmol/L); PKCα, PKCβ, and PKC γ (IC50 = 30 to 42 μmol/L); PKCε, PKCη, and PKCζ (IC50 = 80 to 100 μmol/L). It also can inhibit CaM kinase III (IC50 = 5.3 μmol/L).31Gschwendt M Muller HJ Kielbassa K Zang R Kittstein W Rincke G Marks F Rottlerin, a novel protein kinase inhibitor.Biochem Biophys Res Commun. 1994; 199: 93-98Crossref PubMed Scopus (767) Google Scholar, 32Villalba M Kasibhatla S Genestier L Mahboubi A Green DR Altman A Protein kinase C cooperates with calcineurin to induce fas ligand expression during activation-induced T cell death.J Immunol. 1999; 163: 5813-5819PubMed Google Scholar Cells were preincubated with rottlerin (1.5 μmol/L; IC50 = 3 to 6 μmol/L) for 20 minutes before exposure to HNE, and effects on PKC activity [using detection of phosphorylated (ser) PKC substrate] and on HNE-induced mucin secretion were assessed. As additional controls, the potential role of other PKC isoforms present in these cells was assessed: cells were exposed to the following specific inhibitors for 15 minutes before exposure to HNE and assay for mucin secretion: The PKCα/β inhibitor, Gö 6976 (10 nmol/L; IC50 = 2 ∼ 6 nmol/L);29Martiny-Baron G Kazanietz MG Mischak H Blumberg PM Kochs G Hug H Marme D Schachtele C Selective inhibition of protein kinase C isozymes by the indolocarbazole Go 6976.J Biol Chem. 1993; 268: 9194-9197Abstract Full Text PDF PubMed Google Scholar a PKCζ peptide inhibitor (50 μmol/L; Ser-Ile-Tyr-Arg-Arg-Gly-Ala-Arg-Arg-Trp-Arg-Lys-Leu; IC50 = 10 μmol/L);33Bandyopadhyay G Standaert ML Galloway L Moscat J Farese RV Evidence for involvement of protein kinase C (PKC)-zeta and noninvolvement of diacylglycerol-sensitive PKCs in insulin-stimulated glucose transport in L6 myotubes.Endocrinology. 1997; 138: 4721-4731Crossref PubMed Scopus (210) Google Scholar or a PKCε peptide inhibitor (3 ∼ 300 μmol/L; Glu-Ala-Val-Ser-Leu-Lys-Pro-Thr; IC50 = 80.3 μmol/L).34Johnson JA Gray MO Chen CH Mochly-Rosen D A protein kinase C translocation inhibitor as an isozyme-selective antagonist of cardiac function.J Biol Chem. 1996; 271: 24962-24966Crossref PubMed Scopus (343) Google Scholar, 35Mendez CF Leibiger IB Leibiger B Hoy M Gromada J Berggren PO Bertorello AM Rapid association of protein kinase C-epsilon with insulin granules is essential for insulin exocytosis.J Biol Chem. 2003; 278: 44753-44757Crossref PubMed Scopus (59) Google Scholar Data were expressed as the ratio of treatment to the corresponding vehicle control. Results were evaluated using one-way analysis of variance with Bonferroni posttest correction for multiple comparisons.36Kleinbaum DG Kupper LL Muller KE Applied Regression Analysis and Other Multivariable Methods. PWS-Kent Pub. Co., Boston1988: 341-386Google Scholar A P value of <0.05 was considered significant. All reagents used were tested for cytotoxicity using a Promega Cytotox 96 nonradioactive cytotoxicity assay kit according to the manufacturer's instructions. The data were expressed as the ratio of released lactate dehydrogenase to total lactate dehydrogenase. Released lactate dehydrogenase never exceeded 10% of total lactate dehydrogenase (data not shown) in any of the experiments below. As illustrated in Figure 1, HNE stimulated mucin secretion by NHBE cells. Maximal mucin secretion was elicited after 15 minutes exposure to HNE (Figure 1A) so this time point was chosen for additional experiments. HNE increased mucin secretion in a concentration-dependent manner, with 0.01 to 1.0 μmol/L HNE increasing secretion significantly over vehicle control (Figure 1B). Secretion of major gel-forming mucins, including MUC2, MUC5AC, and MUC5B, was investigated after exposure to HNE. As illustrated in Figure 2, HNE enhanced release of both MUC5AC and MUC5B mucins from NHBE cells in a concentration-dependent manner. Secretion of MUC2 mucin was significantly decreased by HNE. Elastatinal appeared to be the weakest of the three HNE inhibitors used in this study because the highest concentration used, 100 μmol/L, blocked only 50% of HNE enzymatic activity and did not affect HNE-stimulated mucin secretion (Figure 3A). CMK proved to be a more potent HNE enzymatic inhibitor because 50 μmol/L CMK completely blocked the enzymatic activity of 1 μmol/L HNE, whereas lower concentrations partially inhibited HNE activity in a concentration-dependent manner. CMK also showed an inhibitory effect on HNE-stimulated mucin secretion in a concentration-dependent manner w}, number={3}, journal={AMERICAN JOURNAL OF PATHOLOGY}, author={Park, JA and He, F and Martin, LD and Li, YH and Chorley, BN and Adler, KB}, year={2005}, month={Sep}, pages={651–661} } @article{deshmukh_case_wesselkamper_borchers_martin_shertzer_nadel_leikauf_2005, title={Metalloproteinases mediate mucin 5AC expression by epidermal growth factor receptor activation}, volume={171}, ISSN={["1535-4970"]}, DOI={10.1164/rccm.200408-1003OC}, abstractNote={Chronic obstructive pulmonary disease is marked by alveolar enlargement and excess production of airway mucus. Acrolein, a component of cigarette smoke, increases mucin 5AC (MUC5AC), a prevalent airway mucin in NCI-H292 cells by transcriptional activation, but the signal transduction pathways involved in acrolein-induced MUC5AC expression are unknown. Acrolein depleted cellular glutathione at doses of 10 muM or greater, higher than those sufficient (0.03 muM) to increase MUC5AC mRNA, suggesting that MUC5AC expression was independent of oxidative stress. In contrast, acrolein increased MUC5AC mRNA levels by phosphorylating epidermal growth factor receptor (EGFR) and mitogen-activated protein kinase 3/2, or MAPK 3/2(ERK1/2). Pretreating the cells with an EGFR-neutralizing antibody, or a metalloproteinase inhibitor, decreased the acrolein-induced MUC5AC mRNA increase. Small, interfering RNA directed against ADAM17 or MMP9 inhibited the acrolein-induced MUC5AC mRNA increase. Acrolein increased the release and subsequent activation of pro-MMP9. Acrolein increased MMP9 and decreased tissue inhibitor of metalloproteinase 3 (TIMP3), an endogenous inhibitor of ADAM17, transcripts. Together, these data suggest that acrolein induces MUC5AC expression via an initial ligand-dependent activation of EGFR mediated by ADAM17 and MMP9. In addition, a prolonged effect of acrolein may be mediated by altering MMP9 and TIMP3 transcription.}, number={4}, journal={AMERICAN JOURNAL OF RESPIRATORY AND CRITICAL CARE MEDICINE}, author={Deshmukh, HS and Case, LM and Wesselkamper, SC and Borchers, MT and Martin, LD and Shertzer, HG and Nadel, JA and Leikauf, GGD}, year={2005}, month={Feb}, pages={305–314} } @article{lankford_macchione_crews_mckane_akley_martin_2005, title={Modeling the airway epithelium in allergic asthma: Interleukin-13-induced effects in differentiated murine tracheal epithelial cells}, volume={41}, number={7}, journal={In Vitro Cellular & Developmental Biology. Animal}, author={Lankford, S. M. and Macchione, M. and Crews, A. L. and McKane, S. A. and Akley, N. J. and Martin, L. D.}, year={2005}, pages={217–224} } @article{willens_stoskopf_martin_lewbart_2005, title={Viability of glycerol-preserved and cryopreserved anuran skin}, volume={41}, number={09-Aug}, journal={In Vitro Cellular & Developmental Biology. Animal}, author={Willens, S. and Stoskopf, M. K. and Martin, L. D. and Lewbart, G. A.}, year={2005}, pages={258–263} } @article{singer_martin_vargaftig_park_gruber_li_adler_2004, title={A MARCKS-related peptide blocks mucus hypersecretion in a mouse model of asthma}, volume={10}, ISSN={["1546-170X"]}, DOI={10.1038/nm983}, abstractNote={Mucus hypersecretion is a crucial feature of pulmonary diseases such as asthma, chronic bronchitis and cystic fibrosis. Despite much research, there is still no effective therapy for this condition. Recently, we showed that the myristoylated, alanine-rich C-kinase substrate (MARCKS) protein is required for mucus secretion by human bronchial epithelial cells in culture. Having synthesized a peptide corresponding to the N-terminal domain of MARCKS, we now show that the intratracheal instillation of this peptide blocks mucus hypersecretion in a mouse model of asthma. A missense peptide with the same amino acid composition has no effect. Based on quantitative histochemical analysis of the mouse airways, the peptide seems to act by blocking mucus release from goblet cells, possibly by inhibiting the attachment of MARCKS to membranes of intracellular mucin granules. These results support a pivotal role for MARCKS protein, specifically its N-terminal region, in modulating this secretory process in mammalian airways. Intratracheal administration of this MARCKS-related peptide could therapeutically reduce mucus secretion in the airways of human patients with asthma, chronic bronchitis and cystic fibrosis.}, number={2}, journal={NATURE MEDICINE}, author={Singer, M and Martin, LD and Vargaftig, BB and Park, J and Gruber, AD and Li, YH and Adler, KB}, year={2004}, month={Feb}, pages={193–196} } @misc{martin_adler_li_2004, title={Blocking peptide for inflammatory cell secretion}, volume={WO/2003/000027}, number={2004 Sep. 16}, publisher={Washington, DC: U.S. Patent and Trademark Office}, author={Martin, L. D. and Adler, K. B. and Li, Y}, year={2004} } @article{vargaftig_singer_martin_li_adler_2003, title={A myristoylated peptide directed against the N-terminal region of MARCKS protein inhibits mucin secretion in ovalbumin sensitized/challenged mice in vivo.}, volume={167}, journal={American Journal of Respiratory and Critical Care Medicine}, author={Vargaftig, B. and Singer, M. and Martin, L. D. and Li, Y. and Adler, K. B.}, year={2003}, pages={A17} } @article{chorley_martin_crews_li_adler_2003, title={Differential effects of albuterol isomers on normal human bronchial epithelial cells in vitro.}, volume={167}, journal={American Journal of Respiratory and Critical Care Medicine}, author={Chorley, B. N. and Martin, L. D. and Crews, A. C. and Li, Y. and Adler, K. B.}, year={2003}, pages={A205} } @article{krunkosky_martin_fischer_voynow_adler_2003, title={Effects of TNF alpha on expression of ICAM-1 in human airway epithelial cells in vitro: Oxidant-mediated pathways and transcription factors}, volume={35}, ISSN={["1873-4596"]}, DOI={10.1016/S0891-5849(03)00498-2}, abstractNote={We demonstrate that two different cell-permeable antioxidants, pyrrolidine dithiocarbamate (PDTC) and dimethylthiourea (DMTU), inhibit TNFalpha-induced ICAM-1 surface and gene expression in primary cultures of differentiated normal human bronchial epithelial (NHBE) cells. In addition, TNFalpha stimulates binding of nuclear proteins to the nuclear factor kappa beta (NFkappaB) and the CAAT/enhancer binding protein (C/EBP) consensus sites in the ICAM-1 promoter in these cells. Because these transcription factors have been suggested to be oxidant-sensitive and important in ICAM-1 expression, the potential involvement of reactive oxygen species (ROS) in the response to TNFalpha was investigated. Interestingly, neither PDTC nor DMTU altered binding of NFkappaB complexes. In contrast, either the proteasome inhibitor carbobenzoxy-L-leucy-L-leucy-L-leucinal (MG 132) or the IkappaBalpha inhibitor BAY 11-7082 ablated TNFalpha-induced ICAM-1 gene expression and MG132 inhibited TNFalpha-induced NFkappaB complexes. Surprisingly, either PDTC or DMTU inhibited the binding of TNFalpha-enhanced C/EBP complexes to the consensus site directly adjacent to the NFkappaB site. These results suggest that although TNFalpha enhances binding of C/EBP and NFkappaB complexes in NHBE cells, C/EBP binding seems to involve an oxidant-dependent mechanism, whereas activation of NFkappaB complexes utilizes the ubiquitin-proteasome pathway, a mechanism that seems to be unaltered by the presence of antioxidants. Because interference with either signaling pathway abrogates TNFalpha-induced ICAM-1 expression, activation of both complexes seems to be involved in this response to TNFalpha, but this activation occurs via different intracellular pathways.}, number={9}, journal={FREE RADICAL BIOLOGY AND MEDICINE}, author={Krunkosky, TM and Martin, LD and Fischer, BM and Voynow, JA and Adler, KB}, year={2003}, month={Nov}, pages={1158–1167} } @article{park_he_li_martin_adler_2003, title={Human neutrophil elastase provokes release of MUC5B mucin from normal bronchial epithelial cells in vitro via a PKC-dependent mechanism.}, volume={167}, journal={American Journal of Respiratory and Critical Care Medicine}, author={Park, J. A. and He, F. and Li, Y. and Martin, L. D. and Adler, K. B.}, year={2003}, pages={A203} } @article{little_dean_young_mckane_martin_jones_blikslager_2003, title={PI3K signaling is required for prostaglandin-induced mucosal recovery in ischemia-injured porcine ileum}, volume={284}, ISSN={0193-1857 1522-1547}, url={http://dx.doi.org/10.1152/ajpgi.00121.2002}, DOI={10.1152/ajpgi.00121.2002}, abstractNote={ We have previously shown that PGE2 and PGI2 induce recovery of transepithelial resistance (TER) in ischemia-injured porcine ileal mucosa, associated with initial increases in Cl−secretion. We believe that the latter generates an osmotic gradient that stimulates resealing of tight junctions. Because of evidence implicating phosphatidylinositol 3-kinase (PI3K) in regulating tight junction assembly, we postulated that this signaling pathway is involved in PG-induced mucosal recovery. Porcine ileum was subjected to 45 min of ischemia, after which TER was monitored for a 180-min recovery period. Endogenous PG production was inhibited with indomethacin (5 μM). PGE2 (1 μM) and PGI2(1 μM) stimulated recovery of TER, which was inhibited by serosal application of the osmotic agent urea (300 mosmol/kgH2O). The PI3K inhibitor wortmannin (10 nM) blocked recovery of TER in response to PGs or mucosal urea. Immunofluorescence imaging of recovering epithelium revealed that PGs restored occludin and zonula occludens-1 distribution to interepithelial junctions, and this pattern was disrupted by pretreatment with wortmannin. These experiments suggest that PGs stimulate recovery of paracellular resistance via a mechanism involving transepithelial osmotic gradients and PI3K-dependent restoration of tight junction protein distribution. }, number={1}, journal={American Journal of Physiology-Gastrointestinal and Liver Physiology}, publisher={American Physiological Society}, author={Little, Dianne and Dean, Rebecca A. and Young, Karen M. and McKane, Shaun A. and Martin, Linda D. and Jones, Samuel L. and Blikslager, Anthony T.}, year={2003}, month={Jan}, pages={G46–G56} } @article{booth_newcomb_mckane_crews_adler_bonner_martin_2003, title={Proliferation of the airway epithelium in asthma - Are inflammatory cells required?}, volume={123}, ISSN={["0012-3692"]}, DOI={10.1378/chest.123.3_suppl.384S}, abstractNote={Asthma is associated with a T helper type 2 phenotype in which interleukin (IL)-4, IL-5, and IL-13 predominate. In addition, the long-term presence of these inflammatory mediators is thought to lead to airway structural changes that are collectively referred to as airway remodeling. Data from our laboratory, and those of others, have suggested a role for IL-13 in the development of mucous cell hyperplasia that is associated with such remodeling. Others also have suggested a role for inflammatory cells such as neutrophils in mediating this process. Using normal human bronchial epithelial (NHBE) cells differentiated in vitro, we have shown recently that IL-13 (10 ng/mL for 24 h) induces the proliferation of NHBE cells via a mechanism that is dependent on the IL-13-induced release of transforming growth factor (TGF)-α by the epithelial cells. This epithelium-derived TGF-α then acts in an autocrine/paracrine manner to bind the epidermal growth factor receptor (EGFR) on these NHBE cells, enhancing proliferation. Specifically, soluble TGF-α is released by NHBE cells in response to IL-13 exposure (1 h), and the immunohistochemical analysis of cells exposed to IL-13 (after 1 and 4 h) has revealed a lack of membrane-bound TGF-α when compared to control cells. The IL-13-induced proliferative response can be blocked in a concentration-dependent manner by AG1478 (0.1, 1, and 5 μg/mL), which is a specific inhibitor of EGFR tyrosine kinase activity, and is eliminated by neutralizing TGF-α antibodies, while control antibodies (ie, anti-platelet-derived growth factor, epidermal growth factor [EGF], and heparin-binding EGF) have no effect.}, number={3}, journal={CHEST}, author={Booth, BW and Newcomb, DC and McKane, SA and Crews, AL and Adler, KB and Bonner, JC and Martin, LD}, year={2003}, month={Mar}, pages={384S–385S} } @article{booth_adler_martin_2002, title={IL-13 induces intracellular translocation and release of TGF? in human bronchial epithelial cells in vitro.}, volume={165}, journal={American Journal of Respiratory and Critical Care Medicine}, author={Booth, B. W. and Adler, K. B. and Martin, L. D.}, year={2002}, pages={A814} } @article{martin_adler_akley_crews_sharova_2002, title={Secretion-competent mouse tracheal epithelial cell culture from the genetically altered mouse - Pathway analysis via gene array}, volume={121}, DOI={10.1016/S0012-3692(15)35478-7}, abstractNote={The ability to create knockout and transgenic mice with phenotypes mimicking a variety of lung diseases has led to a large body of knowledge detailing the role of various gene products in the development of these diseases. Similarly, the use of well-differentiated human airway epithelial cell cultures has led to an understanding of precise signaling pathways regulating cellular functions such as mucus secretion, adhesion molecule and cytokine expression, and epithelial cell proliferation. The ability to combine these two powerful research approaches lies with creating an in vitro mouse tracheal epithelial (MTE) cell culture system. Here, we report the development of such a primary cell system that maintains morphologic and functional characteristics of the in vivo mouse airway epithelium. Specifically, epithelial cells dissociated from intact mouse tracheas are grown in air/liquid interface culture in defined media with or without serum. Under both conditions, Alcian blue/periodic acid-Schiff–positive mucous cells are observed. In contrast, ciliary development appears to require serum, suggesting that it may be possible to further manipulate this cell culture system to allow precise study of either mucous or ciliated cell development. This cell culture system has been examined to ensure its epithelial nature as indicated by Western blot analyses showing the culture findings to be positive for cytokeratin 5 expression. Using a mouse mucin 5ac-specific antibody to detect secreted protein by enzyme-linked immunosorbent assay, the cultures are found to secrete mucin constitutively and in a stimulated manner in response to known secretagogues (phorbol 12-myristate 13-acetate and 8-Br-cyclic guanosine monophosphate). Although a single trachea yields only 1 cm2 of differentiated culture, our preliminary studies indicate sufficient material can be obtained to perform gene array analyses of control and interleukin-13–exposed MTE cell cultures. Thus, we anticipate use of the MTE cell culture system not only to determine specific signaling pathways important to airway epithelial cell changes during lung disease, but by employing cells from knockout and transgenic mice, we expect to obtain an understanding of how expression of genes controlling these pathways is altered by genetic changes. In this manner, it should be possible to directly interface in vitro experimentation to define precise signaling pathways in airway epithelial cells with in vivo whole animal studies.}, number={3}, journal={Chest}, author={Martin, L. D. and Adler, K. B. and Akley, N. J. and Crews, A. and Sharova, L.}, year={2002}, pages={79S} } @article{booth_bonner_adler_martin_2001, title={Autocrine production of TGFa mediates interleukin 13-induced proliferation of human airway epithelial cells during development of a mucous phenotype in vitro.}, volume={163}, journal={American Journal of Respiratory and Critical Care Medicine}, author={Booth, B. and Bonner, J. C. and Adler, K. B. and Martin, L. D.}, year={2001}, pages={A738} } @article{macchione_akley_adler_martin_2001, title={Differentiation of murine tracheal epithelial cells in vitro.}, volume={163}, journal={American Journal of Respiratory and Critical Care Medicine}, author={Macchione, M. and Akley, N. J. and Adler, K. B. and Martin, L. D.}, year={2001}, pages={A225} } @article{marinakos_anderson_ryan_martin_feldheim_2001, title={Encapsulation, permeability, and cellular uptake characteristics of hollow nanometer-sized conductive polymer capsules}, volume={105}, ISSN={["1520-5207"]}, DOI={10.1021/jp010820d}, abstractNote={The use of nanometer-sized gold particles as templates for the synthesis of hollow poly(pyrrole), poly(N-methylpyrrole), and poly(3-methylthiophene) is described in this paper. Diffusion coefficients of small molecules through the capsule shell were found to vary by almost 3 orders of magnitude depending on the polymer, polymer oxidation state, and counteranion incorporated during polymer synthesis. A small molecule (anthraquinone) and an enzyme (horseradish peroxidase) were trapped inside hollow capsules by attaching them to the template particle prior to polymerization and particle etching. A thin poly(pyrrole) shell protected the enzyme 2 times longer in neat toluene compared to unencapsulated enzyme. Finally, the potential for using conductive polymer nanoparticles for intracellular delivery or diagnostics was examined by administering partice suspensions to 3T3 murine fibroblasts. Particles ranging in size from 25 to 100 nm were engulfed by fibroblasts without compromising cell viability.}, number={37}, journal={JOURNAL OF PHYSICAL CHEMISTRY B}, author={Marinakos, SM and Anderson, MF and Ryan, JA and Martin, LD and Feldheim, DL}, year={2001}, month={Sep}, pages={8872–8876} } @article{li_martin_minnicozzi_greenfeder_fine_pettersen_chorley_adler_2001, title={Enhanced expression of mucin genes in a guinea pig model of allergic asthma}, volume={25}, ISSN={["1535-4989"]}, DOI={10.1165/ajrcmb.25.5.4485}, abstractNote={The ovalbumin (OVA)-sensitized guinea pig is often used as an animal model of asthma and airway hyperreactivity. A characteristic lesion of asthma is excessive production of mucin in the airways. Mechanistic studies of this lesion in guinea pigs have been limited due to lack of mucin gene probes for this species. The aim of the present study was to clone the cDNAs encoding two major airway mucins (Muc2 and Muc5ac) from the guinea pig, and investigate mucin gene expression in lungs of sensitized animals in response to antigen challenge. We isolated and sequenced two cDNA fragments coding for the sequences located within the carboxyl-terminal cysteine-rich region of guinea pig Muc2 and Muc5ac mucins. Comparison of cloned cDNAs with those from other species revealed high degrees of sequence identity and conservation of all cysteine residues in deduced primary sequences. Based on the resultant sequence information, we also designed oligonucleotide primers for specific detection of guinea-pig Muc2 and Muc5ac steady-state mRNA levels via reverse transcriptase/ polymerase chain reaction (RT-PCR). Levels of both Muc2 and Muc5ac mRNA in lungs of OVA-sensitized guinea pigs increased significantly by 30 min after an acute exposure to 0.3% OVA. In addition, levels of eotaxin mRNA also increased in these tissues, but the increases were not significant until 2 h after challenge. Correspondingly, the number of eosinophils in bronchoalveolar lavage fluid did not increase until 4 h postchallenge. Results of these studies suggest that the OVA-sensitized guinea pig responds to allergic challenge with enhanced expression of genes (e.g., eotaxin, Muc2, and Muc5ac) that likely play a role in increased airway inflammation and mucin overproduction, and enhanced mucin gene expression appears to occur before eosinophil infiltration.}, number={5}, journal={AMERICAN JOURNAL OF RESPIRATORY CELL AND MOLECULAR BIOLOGY}, author={Li, YH and Martin, LD and Minnicozzi, M and Greenfeder, S and Fine, J and Pettersen, CA and Chorley, B and Adler, KB}, year={2001}, month={Nov}, pages={644–651} } @inbook{martin_macchione_bonner_booth_akley_adler_2001, title={Interleukin-13 induced mucous cell hyperplasia in airway epithelium.}, booktitle={Cilia and mucus: from development to respiratory disease.}, author={Martin, L. D. and Macchione, M. and Bonner, J. C. and Booth, B. W. and Akley, N. J. and Adler, K. B.}, year={2001}, pages={253–263} } @article{booth_adler_bonner_tournier_martin_2001, title={Interleukin-13 induces proliferation of human airway epithelial cells in vitro via a mechanism mediated by transforming growth factor-alpha}, volume={25}, ISSN={["1535-4989"]}, DOI={10.1165/ajrcmb.25.6.4659}, abstractNote={Remodeling of the airways, as occurs in asthmatic patients, is associated with the continual presence of inflammatory mediators and Th2 cytokines, especially interleukin (IL)-13, during cycles of epithelial injury and repair. In this study, we examined the effect of IL-13 on well-differentiated normal human bronchial epithelial (NHBE) cells maintained in air-liquid interface culture. IL-13 induced proliferation of NHBE cells after 24 h exposure, as reflected by [(3)H]thymidine uptake and cell counts. The effects of IL-13 were mediated through the epidermal growth factor receptor (EGFR), as proliferation was attenuated by AG1478, an EGFR tyrosine kinase inhibitor. Proliferation appeared to be mediated by transforming growth factor (TGF)-alpha, a potent ligand for EGFR, which was released rapidly from NHBE cells in response to IL-13. Neutralizing antibody to TGF-alpha, but not antibodies against other potentially important growth factors (EGF, heparin binding epidermal growth factor-like growth factor [HB-EGF], platelet-derived growth factor [PDGF]), inhibited the mitogenic response to IL-13. This study provides the first experimental evidence that IL-13 can initiate a proliferative response of human airway epithelium in the absence of inflammatory cells or other cell types. The results are consistent with a mechanism whereby IL-13 induces release of TGF-alpha from the epithelial cells, which in turn binds via an autocrine/paracrine-type action to the EGFR, initiating proliferation. IL-13-induced airway remodeling in vivo may involve this epithelium-driven response.}, number={6}, journal={AMERICAN JOURNAL OF RESPIRATORY CELL AND MOLECULAR BIOLOGY}, author={Booth, BW and Adler, KB and Bonner, JC and Tournier, F and Martin, LD}, year={2001}, month={Dec}, pages={739–743} } @article{booth_bonner_akley_macchione_adler_martin_2001, title={Interleukin-13, a mediator of subepithelial fibrosis, enhances growth factor production and proliferation in human airway epithelial cells}, volume={120}, ISSN={["0012-3692"]}, DOI={10.1378/chest.120.1_suppl.S15}, abstractNote={Subepithelial fibrosis is a prominent feature of the remodeled asthmatic airway. The cytokine interleukin (IL)-13, implicated as a mediator in the development of asthma, induces a significant degree of subepithelial fibrosis in the lungs of transgenic mice. Since IL-13 has been shown to exert effects on the airway epithelium, including the development of a mucous phenotype, we have begun to determine whether IL-13 provokes production of factors from the epithelium that could elicit the observed subepithelial fibrotic response. In the studies reported herein, injured airways with regions of regenerating/differentiating cells and regions of normal fully differentiated cells have been mimicked by examining the effects of IL-13 on normal human bronchial epithelial cells during mucociliary differentiation in air/liquid interface culture. Exposure of normal human bronchial epithelial cells to IL-13 resulted in increased production of soluble transforming growth factor (TGF)-a, with the growth factor interacting in an autocrine manner with the epidermal growth factor receptor. Production of soluble TGF-a was very rapid, with a threefold increase observed in response to IL-13 (10 ng/mL) b y1ho fexposure. Continuous exposure to IL-13 throughout the course of mucociliary differentiation (a total of 10 days) resulted in a twofold increase in cell number by day 7 when cells are differentiated. Exposure to IL-13 (10 ng/mL; 24 h) provoked a threefold increase in proliferation once the cells were differentiated, an effect that could be duplicated in differ}, number={1}, journal={CHEST}, author={Booth, B and Bonner, J and Akley, N and Macchione, M and Adler, K and Martin, LD}, year={2001}, month={Jul}, pages={15S–15S} } @article{li_pettersen_martin_adler_2001, title={MARCKS protein interaction with the cellular contractile machinery may regulate mucin secretion by human airway epithelium.}, volume={163}, journal={American Journal of Respiratory and Critical Care Medicine}, author={Li, Y. and Pettersen, C. A. and Martin, L. D. and Adler, K. B.}, year={2001}, pages={A225} } @article{li_martin_spizz_adler_2001, title={MARCKS protein is a key molecule regulating mucin secretion by human airway epithelial cells in vitro}, volume={276}, ISSN={["0021-9258"]}, DOI={10.1074/jbc.M105614200}, abstractNote={Hypersecretion of airway mucin characterizes numerous respiratory diseases. Although diverse pathological stimuli can provoke exocytotic release of mucin from secretory cells of the airway epithelium, mechanisms involved remain obscure. This report describes a new paradigm for the intracellular signaling mechanism regulating airway mucin secretion. Direct evidence is provided that the myristoylated alanine-rich C kinase substrate (MARCKS) is a central regulatory molecule linking secretagogue stimulation at the cell surface to mucin granule release by differentiated normal human bronchial epithelial cells in vitro. Down-regulation of MARCKS expression or disruption of MARCKS function in these cells inhibits the secretory response to subsequent stimulation. The intracellular mechanism controlling this secretory process involves cooperative action of two separate protein kinases, protein kinase C and cGMP-dependent protein kinase. Upon stimulation, activated protein kinase C phosphorylates MARCKS, causing translocation of MARCKS from the plasma membrane to the cytoplasm, where it is then dephosphorylated by a protein phosphatase 2A that is activated by cGMP-dependent protein kinase, and associates with both actin and myosin. Dephosphorylated cytoplasmic MARCKS would also be free to interact with mucin granule membranes and thus could link granules to the contractile cytoskeleton, mediating their movement to the cell periphery and subsequent exocytosis. These findings suggest several novel intracellular targets for pharmacological intervention in disorders involving aberrant secretion of respiratory mucin and may relate to other lesions involving exocytosis of membrane-bound granules in various cells and tissues.}, number={44}, journal={JOURNAL OF BIOLOGICAL CHEMISTRY}, author={Li, YH and Martin, LD and Spizz, G and Adler, KB}, year={2001}, month={Nov}, pages={40982–40990} } @inbook{li_martin_adler_2001, title={MARCKS protein: a potential modulator of airway mucin secretion.}, booktitle={Cilia and mucus: from development to respiratory disease.}, author={Li, Y. and Martin, L. D. and Adler, K. B.}, year={2001}, pages={179–193} } @article{zhang_rice_adler_sannes_martin_gladwell_koo_gray_bonner_2001, title={Vanadium stimulates human bronchial epithelial cells to produce heparin-binding epidermal growth factor-like growth factor - A mitogen for lung fibroblasts}, volume={24}, ISSN={["1044-1549"]}, DOI={10.1165/ajrcmb.24.2.4096}, abstractNote={The bronchial epithelium is a potential source of growth factors that could mediate airway fibrosis during the progression of diseases such as asthma and chronic bronchitis. We report that conditioned medium (CM) from normal human bronchial epithelial cells (NHBECs) contains mitogenic activity for human lung fibroblasts that is blocked by the epidermal growth factor receptor (EGF-R) tyrosine kinase inhibitor AG1478 and by neutralizing antibodies raised against heparin-binding epidermal growth factor-like growth factor (HB-EGF). Neutralizing antibodies against other EGF-R ligands (EGF and transforming growth factor-alpha) or other antibodies against growth factors (platelet-derived growth factors, insulin-like growth factor-1) had no affect on the mitogenic activity of NHBEC-CM. HB-EGF messenger RNA (mRNA) expression in NHBEC was detected by reverse transcriptase/polymerase chain reaction and Northern blot analysis. HB-EGF protein was detected by enzyme-linked immunosorbent assay. Vanadium pentoxide (V2O5), a fibrogenic metal associated with occupational asthma, caused a several-fold increase in HB-EGF mRNA expression and protein, whereas the inert metal titanium dioxide had no effect on HB-EGF expression. V2O5-induced HB-EGF mRNA expression was inhibited by the EGF-R tyrosine kinase inhibitor AG1478, the p38 mitogen-activated protein (MAP) kinase inhibitor SB203580, and the MAP kinase kinase inhibitor PD98059. Finally, HB-EGF induced the production of fibroblast growth factor (FGF)-2 by human lung fibroblasts and anti-FGF-2 antibody partially blocked the mitogenic activity of NHBEC-CM on fibroblasts. These data suggest that HB-EGF is a fibroblast mitogen produced by NHBECs and that induction of an FGF-2 autocrine loop in fibroblasts by HB-EGF accounts for part of this mitogenic activity.}, number={2}, journal={AMERICAN JOURNAL OF RESPIRATORY CELL AND MOLECULAR BIOLOGY}, author={Zhang, LM and Rice, AB and Adler, K and Sannes, P and Martin, L and Gladwell, W and Koo, JS and Gray, TE and Bonner, JC}, year={2001}, month={Feb}, pages={123–131} } @inbook{he_adler_akley_fischer_jiang_krunkosky_martin_2000, title={Air-liquid interface culture systems for exposure of differentiated cells to oxidant stress.}, booktitle={Models and methods in cell signaling and gene expression applications to oxidative stress research.}, author={He, F. and Adler, K. B. and Akley, N. J. and Fischer, B. M. and Jiang, N. and Krunkosky, T. M. and Martin, L. D.}, year={2000}, pages={15–30} } @article{martin_krunkosky_adler_2000, title={Differential generation of intracellular oxidants by TNF? and exogenous superoxide affords distinct expression of IL-6 in human airway epithelium in vitro.}, volume={161}, journal={American Journal of Respiratory and Critical Care Medicine}, author={Martin, L. D. and Krunkosky, T. M. and Adler, K. B.}, year={2000}, pages={A152} } @article{krunkosky_fischer_martin_jones_akley_adler_2000, title={Effects of TNF-alpha on expression of ICAM-1 in human airway epithelial cells in vitro - Signaling pathways controlling surface and gene expression}, volume={22}, ISSN={["1535-4989"]}, DOI={10.1165/ajrcmb.22.6.3925}, abstractNote={Signaling pathways associated with tumor necrosis factor (TNF)- α –induced intercellular adhesion molecule 1 (ICAM-1) surface and gene expression were investigated in well differentiated normal human bronchial epithelial (NHBE) cells in air–liquid interface primary culture. Cells were exposed to human recombinant TNF- α (hrTNF- α ; 0.015 to 150 ng/ml [specific activity, 2.86 × 107 U/mg]). TNF- α enhanced ICAM-1 surface expression (measured by flow cytometry) and steady-state messenger RNA (mRNA) levels (assessed by Northern hybridization) in concentration- and time-dependent manners. TNF- α –induced ICAM-1 surface and gene expression were both blocked by the RNA polymerase II inhibitor actinomycin D (0.1 μ g/ml), and surface expression was attenuated by a neutralizing monoclonal antibody directed against the TNF- α receptor p55 (TNF-RI). The intracellular signaling pathway leading to enhanced expression appeared to involve activation of a phospholipase C that hydrolyzes phosphatidylcholine (PC-PLC) becaus...}, number={6}, journal={AMERICAN JOURNAL OF RESPIRATORY CELL AND MOLECULAR BIOLOGY}, author={Krunkosky, TM and Fischer, BM and Martin, LD and Jones, N and Akley, NJ and Adler, KB}, year={2000}, month={Jun}, pages={685–692} } @article{bonner_zhang_sannes_martin_gladwell_koo_gray_adler_2000, title={Induction of heparin-binding epidermal growth factor (HB-EGF) mRNA in normal human bronchial epithelial cells by metal-induced oxidative stress.}, volume={161}, journal={American Journal of Respiratory and Critical Care Medicine}, author={Bonner, J. C. and Zhang, L. and Sannes, P. and Martin, L. and Gladwell, W. and Koo, J.-S. and Gray, T. and Adler, K.}, year={2000}, pages={A149} } @article{martin_bonner_macchione_booth_akley_adler_2000, title={Interaction of TGF? and EGF receptor mediates IL-13 induced mucous cell hyperplasia in human airway epithelium in vitro.}, volume={161}, journal={American Journal of Respiratory and Critical Care Medicine}, author={Martin, L. D. and Bonner, J. C. and Macchione, M. and Booth, B. and Akley, N. and Adler, K. B.}, year={2000}, pages={A779} } @article{li_martin_adler_2000, title={MARCKS protein: a key intracellular molecule controlling mucin secretion by human airway goblet cells.}, volume={161}, journal={American Journal of Respiratory and Critical Care Medicine}, author={Li, Y. and Martin, L. D. and Adler, K. B.}, year={2000}, pages={A259} } @article{he_martin_adler_2000, title={Mechanisms of human neutrophil elastase-induced mucin hypersecretion by human airway epithelial cells in vitro.}, volume={161}, journal={American Journal of Respiratory and Critical Care Medicine}, author={He, F. and Martin, L. D. and Adler, K. B.}, year={2000}, pages={A155} } @article{adler_li_martin_2000, title={Myristoylated alanine-rich C-kinase substrate protein: A major intracellular regulatory molecule controlling secretion of mucin by human airway goblet cells.}, volume={117}, number={5 Supplement 1}, journal={Chest}, author={Adler, K. B. and Li, Y. and Martin, L. D.}, year={2000}, pages={266S–267S} } @article{jiang_dreher_dye_li_richards_martin_adler_2000, title={Residual oil fly ash induces cytotoxicity and mucin secretion by guinea pig tracheal epithelial cells via an oxidant-mediated mechanism}, volume={163}, ISSN={["0041-008X"]}, DOI={10.1006/taap.1999.8886}, abstractNote={Inhalation of ambient air particulate matter (PM) is associated with pulmonary injury and inflammation. Using primary cultures of guinea pig tracheal epithelial (GPTE) cells as an in vitro model of airway epithelium, we examined effects of exposure to suspensions of six different emission and ambient air PM samples: residual oil fly ash (ROFA) from an electrical power plant; fly ash from a domestic oil burning furnace (DOFA); ambient air dust from St. Louis (STL), Ottawa (OT), and Washington, DC (WDC); and volcanic ash from the eruption of Mount Saint Helens (MSH) in 1980. Effects of these particulates on cell viability (assessed via LDH assay), secretion of mucin (measured by a monoclonal antibody-based ELISA), and steady-state mRNA levels of the mucin gene MUC2 were determined. ROFA was the most toxic of the dusts tested, as it significantly increased LDH release following a 24-h incubation with 50 microg/cm(2) ROFA. ROFA also enhanced MUC2 mRNA after 4-h exposure, and mucin secretion after 8 h. ROFA-induced mucin secretion and cytotoxicity were attenuated by the oxidant scavenger, dimethylthiourea (DMTU). ROFA exposure also depleted cells of glutathione (GSH). Relatedly, depletion of intracellular GSH by treatment of the cells with buthionine sulfoxamine (BSO) also provoked mucin secretion, as well as enhancing the secretory effect of ROFA when the two agents were added together. L-NMA, the nitric oxide synthase (NOS) inhibitor, did not affect ROFA-induced mucin secretion. Of the soluble transition metals in ROFA (nickel, iron, vanadium), only vanadium individually, or combinations of the metals containing vanadium, provoked secretion. The results suggest ROFA enhances mucin secretion and generates toxicity in vitro to airway epithelium via a mechanism(s) involving generation of oxidant stress, perhaps related to depletion of cellular antioxidant capacity. Deleterious effects of inhalation of ROFA in the respiratory tract in vivo may relate to these cellular responses. Vanadium, a component of ROFA, may be important in generating these reactions.}, number={3}, journal={TOXICOLOGY AND APPLIED PHARMACOLOGY}, author={Jiang, NF and Dreher, KL and Dye, JA and Li, YH and Richards, JH and Martin, LD and Adler, KB}, year={2000}, month={Mar}, pages={221–230} } @article{martin_norford_voynow_adler_2000, title={Response of human airway epithelium in vitro to inflammatory mediators - Dependence on the state of cellular differentiation}, volume={117}, ISSN={["0012-3692"]}, DOI={10.1378/chest.117.5_suppl_1.267s}, abstractNote={A irways in patients with COPD or chronic bronchitis contain regions of damaged and regenerating epithelium intermixed with normally differentiated mucociliary areas. Responses of these different regions to the inflammatory milieu present in airways of these individuals may differ, thereby altering further development of additional lesions in the airways. In the studies reported here, we utilized normal human bronchial epithelial (NHBE) cells cultured in an air/liquid interface system as a model of well-differentiated epithelium, and the same cells cultured on plastic and submerged in medium as a model of poorly differentiated, regenerating epithelium. We investigated the responses of these different cell types to inflammatory mediators present in inflamed airways: the cytokine interleukin (IL)-13, human neutrophil elastase (HNE), and “cytomix” (10 ng/mL tumor necrosis factor-a, interferon-g, and IL-1b). Acute exposure to IL-13 (10 ng/mL, 24 h) caused an increase in steady-state messenger RNA (mRNA) for mucin (MUC5AC) in undifferentiated cells, but did not affect MUC5AC expression in differentiated cells. Secretion of mucin and the secondary cytokine, IL-6, were both decreased in differentiated epithelial cell cultures after exposure to IL-13, but no secretory change was observed in undifferentiated cells. By contrast, chronic exposure to IL-13 (10 ng/mL, 8 days) caused an increase in mucin secretion in differentiated airway epithelial cells, and a decrease in undifferentiated cells. In response to HNE, well-differentiated cells increased steady-state levels of MUC5AC mRNA, but undifferentiated cells increased mRNA levels of another mucin gene, MUC4. Finally, the message level of inducible nitric oxide synthase (NOS) was increased by cytomix only in differentiated NHBE cultures. Undifferentiated cells did not express inducible NOS at all, but rather the constitutive forms of NOS, endothelial NOS and brain NOS. These data suggest that the response of the airway epithelium to inflammatory mediators may be markedly different in undifferentiated vs fully differentiated cells, and these responses may play a role in further exacerbation of airway inflammation. In vitro studies utilizing cultured airway epithelial cells must take the state of differentiation of these cells into account when analyzing such responses and extrapolating to the in vivo situation.}, number={5}, journal={CHEST}, author={Martin, LD and Norford, D and Voynow, J and Adler, KB}, year={2000}, month={May}, pages={267S–267S} } @article{li_greenfeder_martin_minnicozzi_voynow_adler_1999, title={Cloning and expression of guinea pig MUC2 and MUC5AC genes.}, volume={159}, journal={American Journal of Respiratory and Critical Care Medicine}, author={Li, Y. and Greenfeder, S. and Martin, L. D. and Minnicozzi, M. and Voynow, J. A. and Adler, K. B.}, year={1999}, pages={A852} } @article{li_he_martin_krunkosky_lincoln_cornwell_adler_1999, title={Myristoylated alanine-rich C kinase substrate (MARCKS) is produced by human airway epithelial cells and is phosphorylated by PKC and PKG.}, volume={159}, journal={American Journal of Respiratory and Critical Care Medicine}, author={Li, Y. and He, F. and Martin, L. D. and Krunkosky, T. M. and Lincoln, T. M. and Cornwell, T. L. and Adler, K. B.}, year={1999}, pages={A723} } @article{adler_martin_voynow_krunkosky_1999, title={TNF? effects on human airway epithelial cells in vitro: molecular mechanisms of enhanced mucin production/secretion and ICAM-I expression.}, volume={8}, journal={404nOtfound}, author={Adler, K. B. and Martin, L. D. and Voynow, J. A. and Krunkosky, T. M.}, year={1999}, pages={S55} } @article{krunkosky_martin_li_adler_1999, title={TNF?-induced ICAM-1 expression in airway epithelium: involvement of IkB?.}, volume={159}, journal={American Journal of Respiratory and Critical Care Medicine}, author={Krunkosky, T. M. and Martin, L. D. and Li, Y. and Adler, K. B.}, year={1999}, pages={A184} } @article{martin_krunkosky_adler_1999, title={Transcription factor activation during oxidant-regulated interleukin-6 expression in normal human bronchial epithelium in vitro.}, volume={159}, journal={American Journal of Respiratory and Critical Care Medicine}, author={Martin, L. D. and Krunkosky, T. M. and Adler, K. B.}, year={1999}, pages={A447} } @article{adler_fischer_martin_voynow_1998, title={Effects of inflammatory mediators and drugs on mucus secretion and mucociliary function.}, volume={149}, journal={404nOtfound}, author={Adler, K. B. and Fischer, B. M. and Martin, L. D. and Voynow, J. A.}, year={1998}, pages={245–248} } @article{martin_krunkosky_voynow_adler_1998, title={Intracellular reactive oxygen and nitrogen species as signaling molecules in airway epithelium.}, volume={106}, journal={Environmental Health Perspectives}, author={Martin, L. D. and Krunkosky, T. M. and Voynow, J. A. and Adler, K. B.}, year={1998}, pages={1197–1203} } @inbook{martin_krunkosky_voynow_adler_1998, title={Oxidant-regulated gene expression in inflammatory lung disease.}, DOI={10.1007/978-1-4419-8634-4_23}, booktitle={Acute respiratory distress syndrome: cellular and molecular mechanisms and clinical management.}, author={Martin, L. D. and Krunkosky, T. M. and Voynow, J. A. and Adler, K. B.}, year={1998}, pages={187–195} } @article{martin_krunkosky_voynow_adler_1998, title={The role of reactive oxygen and nitrogen species in airway epithelial gene expression}, volume={106}, ISSN={["0091-6765"]}, DOI={10.2307/3433986}, abstractNote={The body first encounters deleterious inhaled substances, such as allergens, industrial particles, pollutants, and infectious agents, at the airway epithelium.When this occurs, the epithelium and its resident inflammatory cells respond defensively by increasing production of cytokines, mucus, and reactive oxygen and nitrogen species (ROS/RNS).As inflammation in the airway increases, additional infiltrating cells increase the level of these products.Recent interest has focused on ROS/RNS as potential modulators of the expression of inflammation-associated genes important to the pathogenesis of various respiratory diseases.ROS/RNS appear to play a variety of roles that lead to changes in expression of genes such as interleukin-6 and intercellular adhesion molecule 1.By controlling this regulation, the reactive species can serve as exogenous stimuli, as intercellular signaling molecules, and as modulators of the redox state in epithelial cells.Unraveling the molecular mechanisms affected by ROS/RNS acting in these capacities should aid in the understanding of how stimulated defense mechanisms within the airway can lead to disease.}, journal={ENVIRONMENTAL HEALTH PERSPECTIVES}, author={Martin, LD and Krunkosky, TM and Voynow, JA and Adler, KB}, year={1998}, month={Oct}, pages={1197–1203} } @misc{martin_rochelle_fischer_krunkosky_adler_1997, title={Airway epithelium as an effector of inflammation: molecular regulation of secondary mediators}, volume={10}, ISSN={["0903-1936"]}, DOI={10.1183/09031936.97.10092139}, abstractNote={Deleterious environmental stimuli cause the airway epithelium to respond with increased secretions of mucus, reaction of oxygen/nitrogen species, changes in ciliary beating, and the influx of inflammatory cells. The epithelium is a target for factors released by infiltrating inflammatory cells, and has recently been shown to serve as an effector of such inflammation. Molecular mechanisms regulating production of secondary inflammatory mediators (cytokines, lipid mediators, and reactive oxygen/nitrogen species) have yet to be fully described. This report reviews the production of secondary mediators by epithelial cells and by airway epithelium. Lipid mediators are enzymatically produced by the airway epithelium in response to primary mediators. Molecular mechanisms regulating the production of cyclo-oxygenase, lipoxygenase and prostaglandin synthase are discussed, along with the potential of lipid mediators to produce inflammation. The molecular regulation of nitric oxide production is also described in the context of its role as a signalling molecule in pathways regulating secretion of mucus, ciliary motion, and intercellular adhesion molecule-1 (ICAM-1) expression. The production of cytokines by the airway epithelium is shown to play a role in causing inflammation associated with respiratory diseases. Particular attention is paid to molecular mechanisms governing the expression of tumour necrosis factor-alpha (TNF-alpha), interleukin-6 (IL-6), and interleukin-8 (IL-8).}, number={9}, journal={EUROPEAN RESPIRATORY JOURNAL}, author={Martin, LD and Rochelle, LG and Fischer, BM and Krunkosky, TM and Adler, KB}, year={1997}, month={Sep}, pages={2139–2146} } @article{krunkosky_martin_fischer_adler_1997, title={Mechanisms of TNF?-induced ICAM-1 expression in airway epithelium.}, volume={155}, journal={American Journal of Respiratory and Critical Care Medicine}, author={Krunkosky, T. M. and Martin, L. D. and Fischer, B. M. and Adler, K. B.}, year={1997}, pages={A207} } @article{martin_krunkosky_rochelle_adler_1997, title={TNF? and oxidant-induced expression of Interleukin-6 in normal human bronchial epithelium in vitro.}, volume={155}, journal={American Journal of Respiratory and Critical Care Medicine}, author={Martin, L. D. and Krunkosky, T. M. and Rochelle, L. G. and Adler, K. B.}, year={1997}, pages={A206} } @article{martin_krunkosky_dye_fischer_jiang_rochelle_akley_dreher_adler_1997, title={The role of reactive oxygen and nitrogen species in the response of airway epithelium to particulates}, volume={105}, ISSN={["1552-9924"]}, DOI={10.2307/3433551}, abstractNote={Epidemiologic and occupational studies indicate adverse health effects due to inhalation of particulate air pollutants, but precise biologic mechanisms responsible have yet to be fully established.The tracheobronchial epithelium forms the body's first physiologic barrier to such airborne pollutants, where ciliary movement functions to remove the offending substances caught in the overlying mucus layer.Resident and infiltrating phagocytic cells also function in this removal process.In this paper, we examine the role of reactive oxygen and nitrogen species (ROS/RNS) in the response of airway epithelium to particulates.Some particulates themselves can generate ROS, as can the epithelial cells, in response to appropriate stimulation.In addition, resident macrophages in the airways and the alveolar spaces can release ROS/RNS after phagocytosis of inhaled particles.These macrophages also release large amounts of tumor necrosis factor alpha (TNF-a), a cytokine that can generate responses within the airway epithelium dependent upon intracellular generation of ROS/RNS.As a result, signal transduction pathways are set in motion that may contribute to inflammation and other pathobiology in the airway.Such effects include increased expression of intercellular adhesion molecule 1, interleukin-6, cytosolic and inducible nitric oxide synthase, manganese superoxide dismutase, cytosolic phospholipase A2, and hypersecretion of mucus.Ultimately, ROS/RNS may play a role in the global response of the airway epithelium to particulate pollutants via activation of kinases and transcription factors common to many response genes.Thus, defense mechanisms involved in responding to offending particulates may result in a complex cascade of events that can contribute to airway pathology.}, journal={ENVIRONMENTAL HEALTH PERSPECTIVES}, author={Martin, LD and Krunkosky, TM and Dye, JA and Fischer, BM and Jiang, NF and Rochelle, LG and Akley, NJ and Dreher, KL and Adler, KB}, year={1997}, month={Sep}, pages={1301–1307} } @article{adler_krunkosky_fischer_rochelle_martin_dreher_jiang_dye_1996, title={Role or reactive oxygen and nitrogen species in the response of airway epithelium to particulates.}, volume={6}, journal={Proceedings of the 6th International Meeting of the Toxicology of Natural and Man-Made Fibrous and Non-Fibrous Particles.}, author={Adler, K. B. and Krunkosky, T. M. and Fischer, B. M. and Rochelle, L. G. and Martin, L. D. and Dreher, K. L. and Jiang, N. and Dye, J.}, year={1996}, pages={139} } @article{krunkosky_fischer_martin_adler_1996, title={Tumor necrosis factor alpha (TNF?) provokes expression of intercellular adhesion molecule-1 (ICAM-1) on human bronchial epithelial cells in primary culture.}, volume={153}, journal={American Journal of Respiratory and Critical Care Medicine}, author={Krunkosky, T. M. and Fischer, B. M. and Martin, L. D. and Adler, K. B.}, year={1996}, pages={A27} }