Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Lubna H. Abdullah is active.

Publication


Featured researches published by Lubna H. Abdullah.


The Journal of Physiology | 2007

Coordinated release of nucleotides and mucin from human airway epithelial Calu-3 cells

Silvia M. Kreda; Seiko F. Okada; Catharina van Heusden; Wanda K. O'Neal; Sherif E. Gabriel; Lubna H. Abdullah; C. William Davis; Richard C. Boucher; Eduardo R. Lazarowski

The efficiency of the mucociliary clearance (MCC) process that removes noxious materials from airway surfaces depends on the balance between mucin secretion, airway surface liquid (ASL) volume, and ciliary beating. Effective mucin dispersion into ASL requires salt and water secretion onto the mucosal surface, but how mucin secretion rate is coordinated with ion and, ultimately, water transport rates is poorly understood. Several components of MCC, including electrolyte and water transport, are regulated by nucleotides in the ASL interacting with purinergic receptors. Using polarized monolayers of airway epithelial Calu‐3 cells, we investigated whether mucin secretion was accompanied by nucleotide release. Electron microscopic analyses of Calu‐3 cells identified subapical granules that resembled goblet cell mucin granules. Real‐time confocal microscopic analyses revealed that subapical granules, labelled with FM 1‐43 or quinacrine, were competent for Ca2+‐regulated exocytosis. Granules containing MUC5AC were apically secreted via Ca2+‐regulated exocytosis as demonstrated by combined immunolocalization and slot blot analyses. In addition, Calu‐3 cells exhibited Ca2+‐regulated apical release of ATP and UDP‐glucose, a substrate of glycosylation reactions within the secretory pathway. Neither mucin secretion nor ATP release from Calu‐3 cells were affected by activation or inhibition of the cystic fibrosis transmembrane conductance regulator. In SPOC1 cells, an airway goblet cell model, purinergic P2Y2 receptor‐stimulated increase of cytosolic Ca2+ concentration resulted in secretion of both mucins and nucleotides. Our data suggest that nucleotide release is a mechanism by which mucin‐secreting goblet cells produce paracrine signals for mucin hydration within the ASL.


Journal of Clinical Investigation | 2014

Cystic fibrosis airway secretions exhibit mucin hyperconcentration and increased osmotic pressure

Ashley G. Henderson; Camille Ehre; Brian Button; Lubna H. Abdullah; Li Heng Cai; Margaret W. Leigh; Genevieve DeMaria; Hiro Matsui; Scott H. Donaldson; C. William Davis; John K. Sheehan; Richard C. Boucher; Mehmet Kesimer

The pathogenesis of mucoinfective lung disease in cystic fibrosis (CF) patients likely involves poor mucus clearance. A recent model of mucus clearance predicts that mucus flow depends on the relative mucin concentration of the mucus layer compared with that of the periciliary layer; however, mucin concentrations have been difficult to measure in CF secretions. Here, we have shown that the concentration of mucin in CF sputum is low when measured by immunologically based techniques, and mass spectrometric analyses of CF mucins revealed mucin cleavage at antibody recognition sites. Using physical size exclusion chromatography/differential refractometry (SEC/dRI) techniques, we determined that mucin concentrations in CF secretions were higher than those in normal secretions. Measurements of partial osmotic pressures revealed that the partial osmotic pressure of CF sputum and the retained mucus in excised CF lungs were substantially greater than the partial osmotic pressure of normal secretions. Our data reveal that mucin concentration cannot be accurately measured immunologically in proteolytically active CF secretions; mucins are hyperconcentrated in CF secretions; and CF secretion osmotic pressures predict mucus layer-dependent osmotic compression of the periciliary liquid layer in CF lungs. Consequently, mucin hypersecretion likely produces mucus stasis, which contributes to key infectious and inflammatory components of CF lung disease.


The Journal of Physiology | 2008

Munc13-2-/-baseline secretion defect reveals source of oligomeric mucins in mouse airways

Yunxiang Zhu; Camille Ehre; Lubna H. Abdullah; John K. Sheehan; Michelle G. Roy; Christopher M. Evans; Burton F. Dickey; C. William Davis

Since the airways of control mouse lungs contain few alcian blue/periodic acid–Schiffs (AB/PAS)+ staining ‘goblet’ cells in the absence of an inflammatory stimulus such as allergen sensitization, it was surprising to find that the lungs of mice deficient for the exocytic priming protein Munc13‐2 stain prominently with AB/PAS under control conditions. Purinergic agonists (ATP/UTP) stimulated release of accumulated mucins in the Munc13‐2‐deficient airways, suggesting that the other airway isoform, Munc13‐4, supports agonist‐regulated secretion. Notably, however, not all of the mucins in Munc13‐2‐deficient airways were secreted, suggesting a strict Munc13‐2 priming requirement for a population of secretory granules. AB/PAS+ staining of Munc13‐2‐deficient airways was not caused by an inflammatory, metaplastic‐like response: bronchial–alveolar lavage leucocyte numbers, Muc5ac and Muc5b mRNA levels, and Clara cell ultrastructure (except for increased secretory granule numbers) were all normal. A Muc5b‐specific antibody indicated the presence of this mucin in Clara cells of wildtype (WT) control mice, and increased amounts in Munc13‐2‐deficient mice. Munc13‐2 therefore appears to prime a regulated, baseline secretory pathway, such that Clara cell Muc5b, normally secreted soon after synthesis, accumulates in the gene‐deficient animals, making them stain AB/PAS+. The defective priming phenotype is widespread, as goblet cells of several mucosal tissues appear engorged and Clara cells accumulated Clara cell secretory protein (CCSP) in Munc13‐2‐deficient mice. Additionally, because in the human airways, MUC5AC localizes to the surface epithelium and MUC5B to submucosal glands, the finding that Muc5b is secreted by Clara cells under control conditions may indicate that it is also secreted tonically from human bronchiolar Clara cells.


Infection and Immunity | 2014

Localization of Burkholderia cepacia Complex Bacteria in Cystic Fibrosis Lungs and Interactions with Pseudomonas aeruginosa in Hypoxic Mucus

Ute Schwab; Lubna H. Abdullah; Olivia Perlmutt; Daniel A. Albert; C. William Davis; Roland R. Arnold; James R. Yankaskas; Heiner Neubauer; Scott H. Randell; Richard C. Boucher

ABSTRACT The localization of Burkholderia cepacia complex (Bcc) bacteria in cystic fibrosis (CF) lungs, alone or during coinfection with Pseudomonas aeruginosa, is poorly understood. We performed immunohistochemistry for Bcc and P. aeruginosa bacteria on 21 coinfected or singly infected CF lungs obtained at transplantation or autopsy. Parallel in vitro experiments examined the growth of two Bcc species, Burkholderia cenocepacia and Burkholderia multivorans, in environments similar to those occupied by P. aeruginosa in the CF lung. Bcc bacteria were predominantly identified in the CF lung as single cells or small clusters within phagocytes and mucus but not as “biofilm-like structures.” In contrast, P. aeruginosa was identified in biofilm-like masses, but densities appeared to be reduced during coinfection with Bcc bacteria. Based on chemical analyses of CF and non-CF respiratory secretions, a test medium was defined to study Bcc growth and interactions with P. aeruginosa in an environment mimicking the CF lung. When test medium was supplemented with alternative electron acceptors under anaerobic conditions, B. cenocepacia and B. multivorans used fermentation rather than anaerobic respiration to gain energy, consistent with the identification of fermentation products by high-performance liquid chromatography (HPLC). Both Bcc species also expressed mucinases that produced carbon sources from mucins for growth. In the presence of P. aeruginosa in vitro, both Bcc species grew anaerobically but not aerobically. We propose that Bcc bacteria (i) invade a P. aeruginosa-infected CF lung when the airway lumen is anaerobic, (ii) inhibit P. aeruginosa biofilm-like growth, and (iii) expand the host bacterial niche from mucus to also include macrophages.


Brain Research | 1992

Multiphasic effect of morphine on the release of substance P from rat trigeminal nucleus slices

Heberto Suarez-Roca; Lubna H. Abdullah; John R. Zuniga; Sandra Madison; William Maixner

It is generally accepted that morphine acts presynaptically to inhibit substance P (SP) release from afferent terminals in the trigeminal nucleus. Recent studies, however, provide evidence that opioids produce both inhibitory and excitatory effects on SP release which are concentration- and receptor subtype-dependent. In the present study, we have examined a wide range of morphine concentrations on K(+)-evoked SP release from rat trigeminal nucleus caudalis slices. Immunoreactive SP was measured in perfusates. Morphine produced multiphasic effects on K(+)-evoked SP release without affecting basal release. A very low nanomolar concentration (1 nM) suppressed release, higher nanomolar concentrations (100-300 nM) facilitated release, a low micromolar concentration (3 microM) suppressed release, and a higher micromolar concentration (30 microM) facilitated release. These effects were abolished by opioid receptor blockade with naloxone (30 nM). Thus, morphine produces a complex bi-directional modulation of SP release from TNC which is concentration- and possibly receptor subtype-dependent.


Journal of Immunological Methods | 1991

An efficient enzyme immunoassay for glutamate using glutaraldehyde coupling of the hapten to microtiter plates

Paul Ordronneau; Lubna H. Abdullah; Peter Petrusz

In order to coat microtiter plates for enzyme immunoassays (EIAs), amino acids and other haptens are usually coupled to larger protein molecules. The formation of such conjugates is not always reproducible. This may lead to inconsistent hapten-protein stoichiometries, unfavorable orientation of the hapten on the protein and/or well-to-well variation in the concentration of the available hapten. In the assay described here the excitatory amino acid (EAA) Glu is coupled directly to polystyrene microtiter wells with GA. Each step of the assay was tested for maximum efficiency. The resulting EIA with Glu as a competitor gave excellent reproducibility (coefficient of variation = 5.87%), an EC50 of 2.02 X 10(-5) M and a detection limit of 1.26 X 10(-6) M. This EIA method is generally useful for a variety of antisera to amino acids and small peptides and a wide range of competing substances. It can be used to characterize the conformational requirements for antigen binding, to assay for glutamate or to identify compounds with glutamate-like structure in unknown solutions.


American Journal of Physiology-cell Physiology | 1998

Ca2+ and protein kinase C activation of mucin granule exocytosis in permeabilized SPOC1 cells

C. E. Scott; Lubna H. Abdullah; C. William Davis

Mucin secretion by airway goblet cells is under the control of apical P2Y2, phospholipase C-coupled purinergic receptors. In SPOC1 cells, the mobilization of intracellular Ca2+ by ionomycin or the activation of protein kinase C (PKC) by phorbol 12-myristate 13-acetate (PMA) stimulates mucin secretion in a fully additive fashion [L. H. Abdullah, J. D. Conway, J. A. Cohn, and C. W. Davis. Am. J. Physiol. 273 ( Lung Cell. Mol. Physiol. 17): L201-L210, 1997]. This apparent independence between PKC and Ca2+ in the stimulation of mucin secretion was tested in streptolysin O-permeabilized SPOC1 cells. These cells were fully competent to secrete mucin when Ca2+ was elevated from 100 nM to 3.1 μM for 2 min following permeabilization; the Ca2+EC50 was 2.29 ± 0.07 μM. Permeabilized SPOC1 cells were exposed to PMA or 4α-phorbol at Ca2+ activities ranging from 10 nM to 10 μM. PMA, but not 4α-phorbol, increased mucin release at all Ca2+ activities tested: at 10 nM Ca2+ mucin release was 2.1-fold greater than control and at 4.7 μM Ca2+ mucin release was maximal (3.6-fold increase). PMA stimulated 27% more mucin release at 4.7 μM than at 10 nM Ca2+. Hence, SPOC1 cells possess Ca2+-insensitive, PKC-dependent, and Ca2+-dependent PKC-potentiated pathways for mucin granule exocytosis.


Neuroscience | 1992

Molecular requirements for haften binding to antibodies against glutamate and aspartate

Lubna H. Abdullah; Paul Ordronneau; Peter Petrusz

Molecular requirements for hapten recognition by antibodies raised in rabbits against glutaraldehyde conjugates of L-glutamate and L-aspartate were determined in enzyme immunoassays by measuring the displacement of binding of glutamate and aspartate, respectively, by a large number of selected haptens to two anti-glutamate and two anti-aspartate sera. The results indicate that N-terminal modifications of the amino acids, such as the presence of an N-acetyl or N-carbamyl group or the addition of a second amino acid to form dipeptides with C-terminal glutamate or aspartate, are tolerated to variable degrees, more so by the aspartate than the glutamate antisera. The antibodies possess point-to-point recognition sites for the two carboxyl groups present in both amino acids. Strong shape complementarity between the amino acids and their respective binding sites is suggested by the lack of recognition of the appropriate D stereoisomers by any of the antibodies. Changes in the distance between the two carboxyl groups, or modification, replacement or loss of either or both carboxyl groups, strongly reduce or eliminate binding. Based on these results, we suggest that other antibodies raised to similar conjugates of these amino acids are likely to share similar recognition characteristics. In addition, the results provide a rational background for the evaluation of antibody specificity and the interpretation of results in immunocytochemical studies using antisera to glutamate and aspartate.


American Journal of Respiratory Cell and Molecular Biology | 2013

Inflammation Promotes Airway Epithelial ATP Release via Calcium-Dependent Vesicular Pathways

Seiko F. Okada; Carla M. P. Ribeiro; Juliana I. Sesma; Lucia Seminario-Vidal; Lubna H. Abdullah; Catharina van Heusden; Eduardo R. Lazarowski; Richard C. Boucher

ATP in airway surface liquid (ASL) controls mucociliary clearance functions via the activation of airway epithelial purinergic receptors. However, abnormally elevated ATP levels have been reported in inflamed airways, suggesting that excessive ATP in ASL contributes to airway inflammation. Despite these observations, little is known about the mechanisms of ATP accumulation in the ASL covering inflamed airways. In this study, links between cystic fibrosis (CF)-associated airway inflammation and airway epithelial ATP release were investigated. Primary human bronchial epithelial (HBE) cells isolated from CF lungs exhibited enhanced IL-8 secretion after 6 to 11 days, but not 28 to 35 days, in culture, compared with normal HBE cells. Hypotonic cell swelling-promoted ATP release was increased in 6- to 11-day-old CF HBE cells compared with non-CF HBE cells, but returned to normal values after 28 to 35 days in culture. The exposure of non-CF HBE cells to airway secretions isolated from CF lungs, namely, sterile supernatants of mucopurulent material (SMM), also caused enhanced IL-8 secretion and increased ATP release. The SMM-induced increase in ATP release was sensitive to Ca(2+) chelation and vesicle trafficking/exocytosis inhibitors, but not to pannexin inhibition. Transcript levels of the vesicular nucleotide transporter, but not pannexin 1, were up-regulated after SMM exposure. SMM-treated cultures displayed increased basal mucin secretion, but mucin secretion was not enhanced in response to hypotonic challenge after the exposure of cells to either vehicle or SMM. We propose that CF airway inflammation up-regulates the capacity of airway epithelia to release ATP via Ca(2+)-dependent vesicular mechanisms not associated with mucin granule secretion.


PLOS ONE | 2015

Baseline Goblet Cell Mucin Secretion in the Airways Exceeds Stimulated Secretion over Extended Time Periods, and Is Sensitive to Shear Stress and Intracellular Mucin Stores

Yunxiang Zhu; Lubna H. Abdullah; Sean P. Doyle; Kristine Nguyen; Carla M. P. Ribeiro; Paula A. Vasquez; M. Gregory Forest; Michael I. Lethem; Burton F. Dickey; C. William Davis

Airway mucin secretion studies have focused on goblet cell responses to exogenous agonists almost to the exclusion of baseline mucin secretion (BLMS). In human bronchial epithelial cell cultures (HBECCs), maximal agonist-stimulated secretion exceeds baseline by ~3-fold as measured over hour-long periods, but mucin stores are discharged completely and require 24 h for full restoration. Hence, over 24 h, total baseline exceeds agonist-induced secretion by several-fold. Studies with HBECCs and mouse tracheas showed that BLMS is highly sensitive to mechanical stresses. Harvesting three consecutive 1 h baseline luminal incubations with HBECCs yielded equal rates of BLMS; however, lengthening the middle period to 72 h decreased the respective rate significantly, suggesting a stimulation of BLMS by the gentle washes of HBECC luminal surfaces. BLMS declined exponentially after washing HBECCs (t1/2 = 2.75 h), to rates approaching zero. HBECCs exposed to low perfusion rates exhibited spike-like increases in BLMS when flow was jumped 5-fold: BLMS increased >4 fold, then decreased within 5 min to a stable plateau at 1.5–2-fold over control. Higher flow jumps induced proportionally higher BLMS increases. Inducing mucous hyperplasia in HBECCs increased mucin production, BLMS and agonist-induced secretion. Mouse tracheal BLMS was ~6-fold higher during perfusion, than when flow was stopped. Munc13-2 null mouse tracheas, with their defect of accumulated cellular mucins, exhibited similar BLMS as WT, contrary to predictions of lower values. Graded mucous metaplasia induced in WT and Munc13-2 null tracheas with IL-13, caused proportional increases in BLMS, suggesting that naïve Munc13-2 mouse BLMS is elevated by increased mucin stores. We conclude that BLMS is, [i] a major component of mucin secretion in the lung, [ii] sustained by the mechanical activity of a dynamic lung, [iii] proportional to levels of mucin stores, and [iv] regulated differentially from agonist-induced mucin secretion.

Collaboration


Dive into the Lubna H. Abdullah's collaboration.

Top Co-Authors

Avatar

C. William Davis

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Richard C. Boucher

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Camille Ehre

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

John K. Sheehan

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Mehmet Kesimer

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Yunxiang Zhu

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Peter Petrusz

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Carla M. P. Ribeiro

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Eduardo R. Lazarowski

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Paul Ordronneau

University of North Carolina at Chapel Hill

View shared research outputs
Researchain Logo
Decentralizing Knowledge