Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
PLoS One ; 8(9): e73905, 2013.
Article in English | MEDLINE | ID: mdl-24040112

ABSTRACT

Intestinal current measurements (ICM) from rectal biopsies are a sensitive means to detect cystic fibrosis transmembrane conductance regulator (CFTR) function, but have not been optimized for multicenter use. We piloted multicenter standard operating procedures (SOPs) to detect CFTR activity by ICM and examined key questions for use in clinical trials. SOPs for ICM using human rectal biopsies were developed across three centers and used to characterize ion transport from non-CF and CF subjects (two severe CFTR mutations). All data were centrally evaluated by a blinded interpreter. SOPs were then used across four centers to examine the effect of cold storage on CFTR currents and compare CFTR currents in biopsies from one subject studied simultaneously either at two sites (24 hours post-biopsy) or when biopsies were obtained by either forceps or suction. Rectal biopsies from 44 non-CF and 17 CF subjects were analyzed. Mean differences (µA/cm(2); 95% confidence intervals) between CF and non-CF were forskolin/IBMX=102.6(128.0 to 81.1), carbachol=96.3(118.7 to 73.9), forskolin/IBMX+carbachol=200.9(243.1 to 158.6), and bumetanide=-44.6 (-33.7 to -55.6) (P<0.005, CF vs non-CF for all parameters). Receiver Operating Characteristic curves indicated that each parameter discriminated CF from non-CF subjects (area under the curve of 0.94-0.98). CFTR dependent currents following 18-24 hours of cold storage for forskolin/IBMX, carbachol, and forskolin/IBMX+carbachol stimulation (n=17 non-CF subjects) were 44%, 47.5%, and 47.3%, respectively of those in fresh biopsies. CFTR-dependent currents from biopsies studied after cold storage at two sites simultaneously demonstrated moderate correlation (n=14 non-CF subjects, Pearson correlation coefficients 0.389, 0.484, and 0.533). Similar CFTR dependent currents were detected from fresh biopsies obtained by either forceps or suction (within-subject comparisons, n=22 biopsies from three non-CF subjects). Multicenter ICM is a feasible CFTR outcome measure that discriminates CF from non-CF ion transport, offers unique advantages over other CFTR bioassays, and warrants further development as a potential CFTR biomarker.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Cystic Fibrosis/metabolism , Rectum/metabolism , Adult , Aged , Biopsy , Chlorides/metabolism , Cyclic AMP/metabolism , Cystic Fibrosis/diagnosis , Cystic Fibrosis/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Female , Humans , Male , Middle Aged , ROC Curve , Rectum/pathology , Sodium/metabolism , Young Adult
2.
J Biol Chem ; 286(2): 1381-8, 2011 Jan 14.
Article in English | MEDLINE | ID: mdl-21056985

ABSTRACT

Members of the Anoctamin (Ano)/TMEM16A family have recently been identified as essential subunits of the Ca(2+)-activated chloride channel (CaCC). For example, Ano1 is highly expressed in multiple tissues including airway epithelia, where it acts as an apical conduit for transepithelial Cl(-) secretion and helps regulate lung liquid homeostasis and mucus clearance. However, little is known about the oligomerization of this protein in the plasma membrane. Thus, utilizing mCherry- and eGFP-tagged Ano1 constructs, we conducted biochemical and Förster resonance energy transfer (FRET)-based experiments to determine the quaternary structure of Ano1. FRET and co-immunoprecipitation studies revealed that tagged Ano1 subunits directly associated before they reached the plasma membrane. This association was not altered by changes in cytosolic Ca(2+), suggesting that this is a fixed interaction. To determine the oligomeric structure of Ano1, we performed chemical cross-linking, non-denaturing PAGE, and electromobility shift assays, which revealed that Ano1 exists as a dimer. These data are the first to probe the quaternary structure of Ano1. Understanding the oligomeric nature of Ano1 is an essential step in the development of therapeutic drugs that could be useful in the treatment of cystic fibrosis.


Subject(s)
Calcium/metabolism , Chloride Channels , Cystic Fibrosis/metabolism , Actin Cytoskeleton/physiology , Animals , Anoctamin-1 , Biophysics , Cell Membrane/metabolism , Chloride Channels/chemistry , Chloride Channels/genetics , Chloride Channels/metabolism , Dimerization , Fluorescence Resonance Energy Transfer , Green Fluorescent Proteins/genetics , HEK293 Cells , Humans , Mice , Patch-Clamp Techniques , Protein Interaction Domains and Motifs/physiology , Protein Structure, Quaternary , Protein Structure, Tertiary , Respiratory Mucosa/cytology , Transfection
3.
PLoS Biol ; 7(7): e1000155, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19621064

ABSTRACT

Dysfunction of CFTR in cystic fibrosis (CF) airway epithelium perturbs the normal regulation of ion transport, leading to a reduced volume of airway surface liquid (ASL), mucus dehydration, decreased mucus transport, and mucus plugging of the airways. CFTR is normally expressed in ciliated epithelial cells of the surface and submucosal gland ductal epithelium and submucosal gland acinar cells. Critical questions for the development of gene transfer strategies for CF airway disease are what airway regions require CFTR function and how many epithelial cells require CFTR expression to restore normal ASL volume regulation and mucus transport to CF airway epithelium? An in vitro model of human CF ciliated surface airway epithelium (CF HAE) was used to test whether a human parainfluenza virus (PIV) vector engineered to express CFTR (PIVCFTR) could deliver sufficient CFTR to CF HAE to restore mucus transport, thus correcting the CF phenotype. PIVCFTR delivered CFTR to >60% of airway surface epithelial cells and expressed CFTR protein in CF HAE approximately 100-fold over endogenous levels in non-CF HAE. This efficiency of CFTR delivery fully corrected the basic bioelectric defects of Cl(-) and Na(+) epithelial ion transport and restored ASL volume regulation and mucus transport to levels approaching those of non-CF HAE. To determine the numbers of CF HAE surface epithelial cells required to express CFTR for restoration of mucus transport to normal levels, different amounts of PIVCFTR were used to express CFTR in 3%-65% of the surface epithelial cells of CF HAE and correlated to increasing ASL volumes and mucus transport rates. These data demonstrate for the first time, to our knowledge, that restoration of normal mucus transport rates in CF HAE was achieved after CFTR delivery to 25% of surface epithelial cells. In vivo experimentation in appropriate models will be required to determine what level of mucus transport will afford clinical benefit to CF patients, but we predict that a future goal for corrective gene transfer to the CF human airways in vivo would attempt to target at least 25% of surface epithelial cells to achieve mucus transport rates comparable to those in non-CF airways.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Cystic Fibrosis/metabolism , Epithelial Cells/metabolism , Mucus/metabolism , Respiratory Mucosa/metabolism , Analysis of Variance , Biological Transport/physiology , Cells, Cultured , Chlorides/metabolism , Cystic Fibrosis/genetics , Cystic Fibrosis/pathology , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Gene Expression , Gene Transfer Techniques , Genetic Vectors/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Immunohistochemistry , Microscopy, Fluorescence , Parainfluenza Virus 1, Human/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Respiratory Mucosa/pathology , Reverse Transcriptase Polymerase Chain Reaction , Sodium/metabolism
4.
J Biol Chem ; 284(22): 14875-80, 2009 May 29.
Article in English | MEDLINE | ID: mdl-19363029

ABSTRACT

For almost two decades, it has been postulated that calcium-activated Cl(-) channels (CaCCs) play a role in airway epithelial Cl(-) secretion, but until recently, the molecular identity of the airway CaCC(s) was unknown. Recent studies have unequivocally identified TMEM16A as a glandular epithelial CaCC. We have studied the airway bioelectrics of neonatal mice homozygous for a null allele of Tmem16a (Tmem16a(-/-)) to investigate the role of this channel in Cl(-) secretion in airway surface epithelium. When compared with wild-type tracheas, the Tmem16a(-/-) tracheas exhibited a >60% reduction in purinoceptor (UTP)-regulated CaCC activity. Other members of the Tmem16 gene family, including Tmem16f and Tmem16k, were also detected by reverse transcription-PCR in neonatal tracheal epithelium, suggesting that other family members could be considered as contributing to the small residual UTP response. TMEM16A, however, appeared to contribute little to unstimulated Cl(-) secretion, whereas studies with cystic fibrosis transmembrane conductance regulator (CFTR)-deficient mice and wild-type littermates revealed that unstimulated Cl(-) secretion reflected approximately 50% CFTR activity and approximately 50% non-Tmem16a activity. Interestingly, the tracheas of both the Tmem16a(-/-) and the CFTR(-/-) mice exhibited similar congenital cartilaginous defects that may reflect a common Cl(-) secretory defect mediated by the molecularly distinct Cl(-) channels. Importantly, the residual CaCC activity in Tmem16a(-/-) mice appeared inadequate for normal airway hydration because Tmem16a(-/-) tracheas exhibited significant, neonatal, lumenal mucus accumulation. Our data suggest that TMEM16A CaCC-mediated Cl(-) secretion appears to be necessary for normal airway surface liquid homeostasis.


Subject(s)
Calcium/metabolism , Chloride Channels/metabolism , Trachea/metabolism , Animals , Animals, Newborn , Anoctamin-1 , Chloride Channels/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Electric Conductivity , Mice , Mutation/genetics , Trachea/pathology
5.
J Cell Sci ; 119(Pt 7): 1320-8, 2006 Apr 01.
Article in English | MEDLINE | ID: mdl-16537650

ABSTRACT

A new aspect of research into the pathogenesis of cystic fibrosis (CF) is a genetics-based search for ;modifier genes' that may affect the severity of CF lung disease. Using an alternative, cell biological approach, we show that ITPK1 should be considered a modifier gene. ITPK1 synthesizes an intracellular signal, inositol (3,4,5,6)-tetrakisphosphate [Ins(3,4,5,6)P4]. A bio-activatable, cell-permeable analogue of Ins(3,4,5,6)P4 inhibited Ca2+-dependent secretion of Cl- from polarized monolayers of immortalized mouse tracheal epithelial cells (MTEs). Analysis by high-pressure liquid chromatography showed endogenous Ins(3,4,5,6)P4 levels in CF MTEs were approximately 60% below those in wild-type MTEs (P<0.03). This adaptation, which improves purinergic activation of Ca2+-dependent Cl- secretion in CF MTEs, was exceptionally specific; there was no effect upon the cellular levels of all the other inositol phosphate signals. Real-time PCR provided the explanation: the level of ITPK1 expression in wild-type MTEs was twice as high as that in CF MTEs (P<0.002). The biological impact of this differential gene expression is amplified by ITPK1 being concentrated at the apical membrane of MTEs, which we discovered following confocal immunofluorescence microscopy. Compartmentalization of Ins(3,4,5,6)P4 synthesis adjacent to its site of action will enhance its regulatory capacity.


Subject(s)
Cell Membrane/metabolism , Cell Polarity , Cystic Fibrosis/metabolism , Inositol Phosphates/metabolism , Trachea/cytology , Animals , Cell Line , Cystic Fibrosis/pathology , Disease Models, Animal , Fluorescent Antibody Technique, Direct , Fluorescent Dyes , Indoles , Mice , Microscopy, Confocal , RNA Interference , RNA, Small Interfering/metabolism , Reverse Transcriptase Polymerase Chain Reaction
6.
Am J Respir Cell Mol Biol ; 34(3): 355-63, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16284361

ABSTRACT

Deletion of phenylalanine 508 (deltaF508) accounts for nearly 70% of all mutations that occur in the cystic fibrosis transmembrane conductance regulator (CFTR). The deltaF508 mutation is a class II processing mutation that results in very little or no mature CFTR protein reaching the apical membrane and thus no cAMP-mediated Cl- conductance. Therapeutic strategies have been developed to enhance processing of the defective deltaF508 CFTR molecule so that a functional cAMP-regulated Cl- channel targets to the apical membrane. Sarcoplasmic/endoplasmic reticulum calcium (SERCA) inhibitors, curcumin and thapsigargin, have been reported to effectively correct the CF ion transport defects observed in the deltaF508 CF mice. We investigated the effect of these compounds in human airway epithelial cells to determine if they could induce deltaF508 CFTR maturation, and Cl- secretion. We also used Baby Hamster Kidney cells, heterologously expressing deltaF508 CFTR, to determine if SERCA inhibitors could interfere with the interaction between calnexin and CFTR and thereby correct the deltaF508 CFTR misfolding defect. Finally, at the whole animal level, we tested the ability of curcumin and thapsigargin to (1) induce Cl- secretion and reduce hyperabsorption of Na+ in the nasal epithelia of the deltaF508 mouse in vivo, and (2) induce Cl- secretion in intestine (jejunum and distal colon) and the gallbladder of the deltaF508 CF mouse. We conclude that curcumin and thapsigargin failed to induce maturation of deltaF508 CFTR, or induce Cl- secretion, as measured by biochemical and electrophysiologic techniques in a variety of model systems ranging from cultured cells to in vivo studies.


Subject(s)
Calcium-Transporting ATPases/antagonists & inhibitors , Curcumin/pharmacology , Cystic Fibrosis Transmembrane Conductance Regulator/biosynthesis , Cystic Fibrosis/metabolism , Respiratory Mucosa/drug effects , Thapsigargin/pharmacology , Animals , Bronchi/cytology , Calnexin/metabolism , Cells, Cultured , Chlorides/metabolism , Cricetinae , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Gallbladder/drug effects , Gallbladder/physiology , Humans , Intestines/drug effects , Intestines/physiology , Ion Transport , Mice , Mice, Mutant Strains , Mutation , Protein Folding , Respiratory Mucosa/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases , Sodium/metabolism
7.
Am J Physiol Gastrointest Liver Physiol ; 287(1): G33-41, 2004 Jul.
Article in English | MEDLINE | ID: mdl-14988065

ABSTRACT

The effects of CLCA protein expression on the regulation of Cl(-) conductance by intracellular Ca(2+) and cAMP have been studied previously in nonepithelial cell lines chosen for low backgrounds of endogenous Cl(-) conductance. However, CLCA proteins have been cloned from, and normally function in, differentiated epithelial cells. In this study, we examine the effects of differentiation of the Caco-2 epithelial colon carcinoma cell line on modulation of Cl(-) conductance by pCLCA1 protein expression. Cl(-) transport was measured as (36)Cl(-) efflux, as transepithelial short-circuit currents, and as whole cell patch-clamp current-voltage relations. The rate of (36)Cl(-) efflux and amplitude of currents in patch-clamp studies after the addition of the Ca(2+) ionophore A-23187 were increased significantly by pCLCA1 expression in freshly passaged Caco-2 cells. However, neither endogenous nor pCLCA1-dependent Ca(2+)-sensitive Cl(-) conductance could be detected in 14-day-postpassage cells. In contrast to Ca(2+)-sensitive Cl(-) conductance, endogenous cAMP-dependent Cl(-) conductance does not disappear on Caco-2 differentiation. cAMP-dependent Cl(-) conductance was modulated by pCLCA1 expression in Caco-2 cells, and this modulation was observed in freshly passaged and in mature 14-day-postpassage Caco-2 cultures. pCLCA1 mRNA expression, antigenic pCLCA1 protein epitope expression, and pCLCA1 function as a modulator of cAMP-dependent Cl(-) conductance were retained through differentiation in Caco-2 cells, whereas Ca(2+)-dependent Cl(-) conductance disappeared. We conclude that pCLCA1 expression may increase the sensitivity of preexisting endogenous Cl(-) channels to Ca(2+) and cAMP agonists but apparently lacks inherent Cl(-) channel activity under growth conditions where endogenous channels are not expressed.


Subject(s)
Carcinoma/metabolism , Chloride Channels/metabolism , Colonic Neoplasms/metabolism , Animals , Caco-2 Cells , Calcium/metabolism , Carcinoma/physiopathology , Cell Differentiation , Cell Polarity , Cellular Senescence , Chlorides/metabolism , Colonic Neoplasms/physiopathology , Cyclic AMP/metabolism , Electric Conductivity , Humans , Swine , Time Factors
8.
Biochem Biophys Res Commun ; 298(4): 531-6, 2002 Nov 08.
Article in English | MEDLINE | ID: mdl-12408984

ABSTRACT

Members of the CLCA protein family are expressed in airway and intestinal epithelium, where they may participate in secretory activity as mediators of chloride conductance. A calcium-dependent chloride conductance has been observed upon expression of CLCA proteins in non-epithelial cell lines. The pCLCA1 gene, cloned in our laboratory, codes for a product containing a unique A-kinase consensus acceptor site not found in other CLCA proteins. Calcium-dependent, but not cAMP-dependent, chloride conductance increased when pCLCA1 was expressed in NIH/3T3 fibroblasts. We transfected the Caco-2 human colon carcinoma cell line with pCLCA1 to investigate the regulation of CLCA-associated chloride conductance in this differentiated epithelial cell line. Expression of pCLCA1 in the Caco-2 cell line enhanced cAMP-responsive 36Cl efflux, short circuit current, and whole cell chloride current in these cells. This cAMP-dependent chloride conductance was localized to the apical membrane of polarized Caco-2 cells.


Subject(s)
Chloride Channels/physiology , Chlorides/physiology , Cyclic AMP/physiology , Base Sequence , Caco-2 Cells , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , DNA Primers , Humans , Patch-Clamp Techniques , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
9.
J Gen Physiol ; 120(3): 407-18, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12198094

ABSTRACT

Two Cl(-) conductances have been described in the apical membrane of both human and murine proximal airway epithelia that are thought to play predominant roles in airway hydration: (1) CFTR, which is cAMP regulated and (2) the Ca(2+)-activated Cl(-) conductance (CaCC) whose molecular identity is uncertain. In addition to second messenger regulation, cross talk between these two channels may also exist and, whereas CFTR is absent or defective in cystic fibrosis (CF) airways, CaCC is preserved, and may even be up-regulated. Increased CaCC activity in CF airways is controversial. Hence, we have investigated the effects of CFTR on CaCC activity and have also assessed the relative contributions of these two conductances to airway surface liquid (ASL) height (volume) in murine tracheal epithelia. We find that CaCC is up-regulated in intact murine CF tracheal epithelia, which leads to an increase in UTP-mediated Cl(-)/volume secretion. This up-regulation is dependent on cell polarity and is lost in nonpolarized epithelia. We find no role for an increased electrical driving force in CaCC up-regulation but do find an increased Ca(2+) signal in response to mucosal nucleotides that may contribute to the increased Cl(-)/volume secretion seen in intact epithelia. CFTR plays a critical role in maintaining ASL height under basal conditions and accordingly, ASL height is reduced in CF epithelia. In contrast, CaCC does not appear to significantly affect basal ASL height, but does appear to be important in regulating ASL height in response to released agonists (e.g., mucosal nucleotides). We conclude that both CaCC and the Ca(2+) signal are increased in CF airway epithelia, and that they contribute to acute but not basal regulation of ASL height.


Subject(s)
Calcium/physiology , Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Respiratory Mucosa/physiology , Animals , Calcium/pharmacology , Cell Line , Chloride Channels/physiology , Cystic Fibrosis Transmembrane Conductance Regulator/deficiency , In Vitro Techniques , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Respiratory Mucosa/drug effects
10.
Am J Physiol Cell Physiol ; 283(2): C412-21, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12107050

ABSTRACT

The regulatory behavior, inhibitor sensitivity, and properties of the whole cell chloride conductance observed in cells expressing the cDNA coding for a chloride conductance mediator isoform of the CLCA gene family, pCLCA1, have been studied. Common C-kinase consensus phosphorylation sites between pCLCA1 and the closely related human isoform hCLCA1 are consistent with a role for calcium in channel activation. Both channels are activated rapidly on exposure to the calcium ionophore ionomycin. Direct involvement of calcium in the activation of pCLCA1 was supported by the finding that treatment with the intracellular calcium chelator 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid-AM reduced the rate of chloride efflux from NIH/3T3 cells expressing the pCLCA1 channel. No combination of A-kinase activators used was effective in activating chloride efflux via this channel despite the presence of a unique strong A-kinase consensus site in pCLCA1. Notable differences of pCLCA1 from the reported properties of CLCA family members include the failure of phorbol 12-myristate 13-acetate to activate chloride efflux in cells expressing pCLCA1 and a lack of inhibition of chloride efflux from these cells after treatment with DIDS or dithiothreitol. However, selected inhibitors of anionic conductance inhibited pCLCA1-dependent anion efflux. The electrogenic nature of the ionomycin-dependent efflux of chloride from cells expressing pCLCA1 was confirmed by detection of outwardly rectifying chloride current and inhibition of this current by chloride conductance inhibitors in a whole cell patch-clamp study.


Subject(s)
Chloride Channels/physiology , Egtazic Acid/analogs & derivatives , 3T3 Cells , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , Animals , Anions/metabolism , Calcium/physiology , Chelating Agents/pharmacology , Chloride Channels/antagonists & inhibitors , Chloride Channels/genetics , Chlorides/antagonists & inhibitors , Chlorides/physiology , Consensus Sequence , Egtazic Acid/pharmacology , Electric Conductivity , Ionomycin/pharmacology , Ionophores/pharmacology , Mice , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/physiology , Swine , Tetradecanoylphorbol Acetate/pharmacology
11.
J Biol Chem ; 277(10): 8338-45, 2002 Mar 08.
Article in English | MEDLINE | ID: mdl-11756432

ABSTRACT

The epithelial sodium channel (ENaC) constitutes the rate-limiting step for sodium absorption across airway epithelia, which in turn regulates airway surface liquid (ASL) volume and the efficiency of mucociliary clearance. This role in ASL volume regulation suggests that ENaC activity is influenced by local factors rather than systemic signals indicative of total body volume homeostasis. Based on reports that ENaC may be regulated by extracellular serine protease activity in Xenopus and mouse renal epithelia, we sought to identify proteases that serve similar functions in human airway epithelia. Homology screening of a human airway epithelial cDNA library identified two trypsin-like serine proteases (prostasin and TMPRSS2) that, as revealed by in situ hybridization, are expressed in airway epithelia. Functional studies in the Xenopus oocyte expression system demonstrated that prostasin increased ENaC currents 60--80%, whereas TMPRSS2 markedly decreased ENaC currents and protein levels. Studies of primary nasal epithelial cultures in Ussing chambers revealed that inhibition of endogenous serine protease activity with aprotinin markedly decreased ENaC-mediated currents and sensitized the epithelia to subsequent channel activation by exogenous trypsin. These data, therefore, suggest that protease-mediated regulation of sodium absorption is a function of human airway epithelia, and prostasin is a likely candidate for this activity.


Subject(s)
Gene Expression Regulation , Nasal Mucosa/metabolism , Serine Endopeptidases/chemistry , Serine Endopeptidases/metabolism , Sodium Channels/metabolism , Adult , Age Factors , Amino Acid Sequence , Amino Acids/chemistry , Animals , Blotting, Western , Cell Line , Cells, Cultured , Cloning, Molecular , DNA, Complementary/metabolism , Electrophysiology , Epithelial Sodium Channels , Gene Library , Humans , In Situ Hybridization , Lung/metabolism , Mice , Middle Aged , Molecular Sequence Data , Nasal Mucosa/cytology , Oocytes/metabolism , Protein Binding , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Serine Endopeptidases/pharmacology , Trypsin , Xenopus
SELECTION OF CITATIONS
SEARCH DETAIL
...