Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters










Publication year range
1.
Am J Physiol Cell Physiol ; 322(4): C591-C604, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35196166

ABSTRACT

Primary airway epithelial cells (pAECs) cultivated at air-liquid interface (ALI) conditions are widely used as surrogates for human in vivo epithelia. To extend the proliferative capacity and to enable serially passaging of pAECs, conditional reprogramming (cr) has been employed in recent years. However, ALI epithelia derived from cr cells often display functional changes with increasing passages. This highlights the need for thorough validation of the ALI cultures for the respective application. In our study, we evaluated the use of serially passaged cr nasal epithelial cells (crNECs) as a model to study SARS-CoV-2 infection and effects on ion and water transport. NECs were obtained from healthy individuals and cultivated as ALI epithelia derived from passages 1, 2, 3, and 5. We compared epithelial differentiation, ion and water transport, and infection with SARS-CoV-2 between passages. Our results show that epithelia maintained major differentiation characteristics and physiological ion and water transport properties through all passages. However, the frequency of ciliated cells, short circuit currents reflecting epithelial Na+ channel (ENaC) and cystic fibrosis transmembrane conductance regulator (CFTR) activity and expression of aquaporin 3 and 5 decreased gradually over passages. crNECs also expressed SARS-CoV-2 receptors angiotensin converting enzyme 2 (ACE2) and transmembrane serin2 protease 2 (TMPRSS2) across all passages and allowed SARS-CoV-2 replication in all passages. In summary, we provide evidence that passaged crNECs provide an appropriate model to study SARS-CoV-2 infection and also epithelial transport function when considering some limitations that we defined herein.


Subject(s)
COVID-19 , Cell Differentiation , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Epithelial Cells/metabolism , Humans , Infant, Newborn , SARS-CoV-2
2.
Pflugers Arch ; 473(2): 287-311, 2021 02.
Article in English | MEDLINE | ID: mdl-33386991

ABSTRACT

TGF-ß1 is a major mediator of airway tissue remodelling during atopic asthma and affects tight junctions (TJs) of airway epithelia. However, its impact on TJs of ciliated epithelia is sparsely investigated. Herein we elaborated effects of TGF-ß1 on TJs of primary human bronchial epithelial cells. We demonstrate that TGF-ß1 activates TGF-ß1 receptors TGFBR1 and TGFBR2 resulting in ALK5-mediated phosphorylation of SMAD2. We observed that TGFBR1 and -R2 localize specifically on motile cilia. TGF-ß1 activated accumulation of phosphorylated SMAD2 (pSMAD2-C) at centrioles of motile cilia and at cell nuclei. This triggered an increase in paracellular permeability via cellular redistribution of claudin 3 (CLDN3) from TJs into cell nuclei followed by disruption of epithelial integrity and formation of epithelial lesions. Only ciliated cells express TGF-ß1 receptors; however, nuclear accumulations of pSMAD2-C and CLDN3 redistribution were observed with similar time course in ciliated and non-ciliated cells. In summary, we demonstrate a role of motile cilia in TGF-ß1 sensing and showed that TGF-ß1 disturbs TJ permeability of conductive airway epithelia by redistributing CLDN3 from TJs into cell nuclei. We conclude that the observed effects contribute to loss of epithelial integrity during atopic asthma.


Subject(s)
Bronchi/drug effects , Cilia/drug effects , Claudin-3/metabolism , Epithelial Cells/drug effects , Tight Junctions/drug effects , Transforming Growth Factor beta1/pharmacology , Bronchi/metabolism , Cells, Cultured , Cilia/metabolism , Claudin-3/genetics , Electric Impedance , Epithelial Cells/metabolism , Humans , Permeability , Phosphorylation , Protein Transport , Receptor, Transforming Growth Factor-beta Type I/agonists , Receptor, Transforming Growth Factor-beta Type I/metabolism , Receptor, Transforming Growth Factor-beta Type II/agonists , Receptor, Transforming Growth Factor-beta Type II/metabolism , Signal Transduction , Smad2 Protein/metabolism , Tight Junctions/genetics , Tight Junctions/metabolism
3.
Cells ; 11(1)2021 12 24.
Article in English | MEDLINE | ID: mdl-35011607

ABSTRACT

The lamellar body (LB) of the alveolar type II (ATII) cell is a lysosome-related organelle (LRO) that contains surfactant, a complex mix of mainly lipids and specific surfactant proteins. The major function of surfactant in the lung is the reduction of surface tension and stabilization of alveoli during respiration. Its lack or deficiency may cause various forms of respiratory distress syndrome (RDS). Surfactant is also part of the innate immune system in the lung, defending the organism against air-borne pathogens. The limiting (organelle) membrane that encloses the LB contains various transporters that are in part responsible for translocating lipids and other organic material into the LB. On the other hand, this membrane contains ion transporters and channels that maintain a specific internal ion composition including the acidic pH of about 5. Furthermore, P2X4 receptors, ligand gated ion channels of the danger signal ATP, are expressed in the limiting LB membrane. They play a role in boosting surfactant secretion and fluid clearance. In this review, we discuss the functions of these transporting pathways of the LB, including possible roles in disease and as therapeutic targets, including viral infections such as SARS-CoV-2.


Subject(s)
COVID-19/metabolism , Ion Channels/metabolism , Lamellar Bodies/metabolism , Lung/metabolism , Membrane Transport Proteins/metabolism , Pulmonary Surfactants/metabolism , COVID-19/virology , Humans , Lung/virology , Organelles/metabolism , Organelles/virology , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/virology , SARS-CoV-2/physiology
4.
Cell Signal ; 65: 109421, 2020 01.
Article in English | MEDLINE | ID: mdl-31536749

ABSTRACT

The pulmonary epithelium separates the gaseous intraluminal space of the airways and the aqueous interstitium. This compartimentalization is required for appropriate lung function, it is established during perinatal periods and can be disturbed in lung edema. Herein we elaborated the impact of the air-liquid interface (ALI) on the function of the pulmonary epithelium. We used NCI-H441 epithelia as a well-established and characterized model of distal airway epithelia, which were cultivated either at ALI or (at submerged conditions) at liquid-liquid interface conditions (LLI). Our study revealed that paracellular permeability was increased and claudin 1 (CLDN1) expression levels were reduced under LLI conditions. This was accompanied by elevated c-FOS, c-JUN and retinoic acid receptor α (RARA) expression, as well as cellular retinoic acid (RA) content. Exposure of epithelia to RA derivatives of ALI cultivated epithelia mimicked effects of LLI. The increase in RA content was in line with the identified upregulation of retinoic acid anabolizing enzymes ALDH1A3 and DHRS3. CLDN1 promoter analysis revealed c-FOS and c-JUN as activating transcription factors, whereas activation of RARA reduced CLDN1 promoter activity. We then concluded that ALI/LLI dependent modulation of CLDN1 expression and TJ permeability is under the control of RA synthesis. Activation of RARA results in an inhibition of c-FOS/c-JUN dependent CLDN1 promoter activation and increased TJ permeability. Our results underscore RA signalling as a pivotal mechanism in adjusting TJ properties, which could play a role during birth when the lung changes from LLI to ALI conditions.


Subject(s)
Claudin-1/metabolism , Lung/metabolism , Respiratory Mucosa/drug effects , Tight Junctions/drug effects , Tretinoin/pharmacology , Aldehyde Oxidoreductases/metabolism , Claudin-1/genetics , HEK293 Cells , Humans , Lung/drug effects , Lung/physiology , Permeability/drug effects , Promoter Regions, Genetic , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/metabolism , Respiratory Mucosa/metabolism , Respiratory Mucosa/physiology , Retinoic Acid Receptor alpha/genetics , Retinoic Acid Receptor alpha/metabolism , Signal Transduction , Tight Junctions/metabolism , Tight Junctions/physiology , Transcriptional Activation/drug effects , Tretinoin/metabolism , Up-Regulation
5.
Int J Mol Sci ; 20(13)2019 Jun 30.
Article in English | MEDLINE | ID: mdl-31262043

ABSTRACT

Interleukin-13 (IL-13) drives symptoms in asthma with high levels of T-helper type 2 cells (Th2-cells). Since tight junctions (TJ) constitute the epithelial diffusion barrier, we investigated the effect of IL-13 on TJ in human tracheal epithelial cells. We observed that IL-13 increases paracellular permeability, changes claudin expression pattern and induces intracellular aggregation of the TJ proteins zonlua occludens protein 1, as well as claudins. Furthermore, IL-13 treatment increases expression of ubiquitin conjugating E2 enzyme UBE2Z. Co-localization and proximity ligation assays further showed that ubiquitin and the proteasomal marker PSMA5 co-localize with TJ proteins in IL-13 treated cells, showing that TJ proteins are ubiquitinated following IL-13 exposure. UBE2Z upregulation occurs within the first day after IL-13 exposure. Proteasomal aggregation of ubiquitinated TJ proteins starts three days after IL-13 exposure and transepithelial electrical resistance (TEER) decrease follows the time course of TJ-protein aggregation. Inhibition of JAK/STAT signaling abolishes IL-13 induced effects. Our data suggest that that IL-13 induces ubiquitination and proteasomal aggregation of TJ proteins via JAK/STAT dependent expression of UBE2Z, resulting in opening of TJs. This may contribute to barrier disturbances in pulmonary epithelia and lung damage of patients with inflammatory lung diseases.


Subject(s)
Epithelial Cells/metabolism , Interleukin-13/pharmacology , Tight Junctions/metabolism , Trachea/metabolism , Cells, Cultured , Epithelial Cells/drug effects , Humans , Janus Kinases/metabolism , Proteasome Endopeptidase Complex/metabolism , STAT Transcription Factors/metabolism , Tight Junctions/drug effects , Trachea/cytology , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitination
6.
Pflugers Arch ; 471(4): 519-532, 2019 04.
Article in English | MEDLINE | ID: mdl-30397774

ABSTRACT

The lung is the interface between air and blood where the exchange of oxygen and carbon dioxide occurs. The surface liquid that is directly exposed to the gaseous compartment covers both conducting airways and respiratory zone and forms the air-liquid interface. The barrier that separates this lining fluid of the airways and alveoli from the extracellular compartment is the pulmonary epithelium. The volume of the lining fluid must be kept in a range that guarantees an appropriate gas exchange and other functions, such as mucociliary clearance. It is generally accepted that this is maintained by balancing resorptive and secretory fluid transport across the pulmonary epithelium. Whereas osmosis is considered as the exclusive principle of fluid transport in the airways, filtration may contribute to alveolar fluid accumulation under pathologic conditions. Aquaporins (AQP) facilitate water flux across cell membranes, and as such, they provide a transcellular route for water transport across epithelia. However, their contribution to near-isosmolar fluid conditions in the lung still remains elusive. Herein, we discuss the role of AQPs in the lung with regard to fluid homeostasis across the respiratory epithelium.


Subject(s)
Aquaporins/metabolism , Lung/metabolism , Animals , Biological Transport/physiology , Cell Membrane/metabolism , Humans , Pulmonary Alveoli/metabolism , Respiratory Mucosa/metabolism , Water/metabolism
7.
Am J Physiol Lung Cell Mol Physiol ; 316(1): L58-L70, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30358443

ABSTRACT

Mucus clearance provides an essential innate defense mechanism to keep the airways and lungs free of particles and pathogens. Baseline and stimulated mucin secretion from secretory airway epithelial cells need to be tightly regulated to prevent mucus hypersecretion and mucus plugging of the airways. It is well established that extracellular ATP is a potent stimulus for regulated mucus secretion. Previous studies revealed that ATP acts via metabotropic P2Y2 purinoreceptors on goblet cells. Extracellular ATP, however, is also a potent agonist for ionotropic P2X purinoreceptors. Expression of several P2X isoforms has been reported in airways, but cell type-specific expression and the function thereof remained elusive. With this study, we now provide evidence that P2X4 is the predominant P2X isoform expressed in secretory airway epithelial cells. After IL-13 treatment of either human primary tracheal epithelial cells or mice, P2X4 expression is upregulated in vitro and in vivo under conditions of chronic inflammation, mucous metaplasia, and hyperplasia. Upregulation of P2X4 is strongest in MUC5AC-positive goblet cells. Moreover, activation of P2X4 by extracellular ATP augments intracellular Ca2+ signals and mucin secretion, whereas Ca2+ signals and mucin secretion are dampened by inhibition of P2X4 receptors. These data provide new insights into the purinergic regulation of mucin secretion and add to the emerging picture that P2X receptors modulate exocytosis of large secretory organelles and secretion of macromolecular vesicle cargo.


Subject(s)
Calcium Signaling , Goblet Cells/metabolism , Mucins/metabolism , Receptors, Purinergic P2X4/metabolism , Up-Regulation , Adenosine Triphosphate/pharmacology , Goblet Cells/pathology , Humans , Inflammation/metabolism , Inflammation/pathology
8.
J Physiol ; 596(20): 4893-4907, 2018 10.
Article in English | MEDLINE | ID: mdl-30144063

ABSTRACT

KEY POINTS: Re-sensitization of P2X4 receptors depends on a protonation/de-protonation cycle Protonation and de-protonation of the receptors is achieved by internalization and recycling of P2X4 receptors via acidic compartments Protonation and de-protonation occurs at critical histidine residues within the extracellular loop of P2X4 receptors Re-sensitization is blocked in the presence of the receptor agonist ATP ABSTRACT: P2X4 receptors are members of the P2X receptor family of cation-permeable, ligand-gated ion channels that open in response to the binding of extracellular ATP. P2X4 receptors are implicated in a variety of biological processes, including cardiac function, cell death, pain sensation and immune responses. These physiological functions depend on receptor activation on the cell surface. Receptor activation is followed by receptor desensitization and deactivation upon removal of ATP. Subsequent re-sensitization is required to return the receptor into its resting state. Desensitization and re-sensitization are therefore crucial determinants of P2X receptor signal transduction and responsiveness to ATP. However, the molecular mechanisms controlling desensitization and re-sensitization are not fully understood. In the present study, we provide evidence that internalization and recycling via acidic compartments is essential for P2X4 receptor re-sensitization. Re-sensitization depends on a protonation/de-protonation cycle of critical histidine residues within the extracellular loop of P2X4 receptors that is mediated by receptor internalization and recycling. Interestingly, re-sensitization under acidic conditions is completely revoked by receptor agonist ATP. Our data support the physiological importance of the unique subcellular distribution of P2X4 receptors that is predominantly found within acidic compartments. Based on these findings, we suggest that recycling of P2X4 receptors regulates the cellular responsiveness in the sustained presence of ATP.


Subject(s)
Receptors, Purinergic P2X4/metabolism , Adenosine Triphosphate/metabolism , Binding Sites , HEK293 Cells , HeLa Cells , Humans , Protein Transport , Protons , Receptors, Purinergic P2X4/chemistry , Signal Transduction
9.
FASEB J ; 32(8): 4600-4611, 2018 08.
Article in English | MEDLINE | ID: mdl-29543530

ABSTRACT

The release of pulmonary surfactant by alveolar type II (ATII) cells is essential for lowering surface tension at the respiratory air-liquid interface, stabilizing the lungs against physical forces tending to alveolar collapse. Hydrophobic surfactant protein (SP)-B ensures the proper packing of newly synthesized surfactant particles, promotes the formation of the surface active film at the alveolar air-liquid interface and maintains its proper structure along the respiratory dynamics. We report that membrane-associated SP-B efficiently induces secretion of pulmonary surfactant by ATII cells, at the same level as potent secretagogues such as ATP. The presence in the extracellular medium of lipid-protein complexes containing SP-B activates the P2Y2 purinergic signaling pathway that ultimately triggers exocytosis of lamellar bodies by ATII cells. Our data suggest that SP-B prompts Ca2+-dependent surfactant secretion via ATP release from ATII cells. This result implies that SP-B is not only an essential component for the biophysical function of surfactant but is also a central element in the alveolar homeostasis by eliciting autocrine and paracrine cell stimulation.-Martínez-Calle, M., Olmeda, B., Dietl, P., Frick, M., Pérez-Gil, J. Pulmonary surfactant protein SP-B promotes exocytosis of lamellar bodies in alveolar type II cells.


Subject(s)
Exocytosis/physiology , Lung/metabolism , Lung/physiology , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/physiology , Pulmonary Surfactant-Associated Protein B/metabolism , Pulmonary Surfactants/metabolism , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/physiology , Animals , Calcium/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Purinergic P2Y2/metabolism , Signal Transduction/physiology , Swine
10.
ACS Nano ; 12(2): 1188-1202, 2018 02 27.
Article in English | MEDLINE | ID: mdl-29357226

ABSTRACT

Silver nanoparticles (AgNPs) are microbicidal agents which could be potentially used as an alternative to antivirals to treat human infectious diseases, especially influenza virus infections where antivirals have generally proven unsuccessful. However, concerns about the use of AgNPs on humans arise from their potential toxicity, although mechanisms are not well-understood. We show here, in the context of an influenza virus infection of lung epithelial cells, that AgNPs down-regulated influenza induced CCL-5 and -IFN-ß release (two cytokines important in antiviral immunity) through RIG-I inhibition, while enhancing IL-8 production, a cytokine important for mobilizing host antibacterial responses. AgNPs activity was independent of coating and was not observed with gold nanoparticles. Down-stream analysis indicated that AgNPs disorganized the mitochondrial network and prevented the antiviral IRF-7 transcription factor influx into the nucleus. Importantly, we showed that the modulation of RIG-I-IRF-7 pathway was concomitant with inhibition of either classical or alternative autophagy (ATG-5- and Rab-9 dependent, respectively), depending on the epithelial cell type used. Altogether, this demonstration of a AgNPs-mediated functional dichotomy (down-regulation of IFN-dependent antiviral responses and up-regulation of IL-8-dependent antibacterial responses) may have practical implications for their use in the clinic.


Subject(s)
Antiviral Agents/pharmacology , Epithelial Cells/drug effects , Lung/drug effects , Metal Nanoparticles/chemistry , Mitochondria/drug effects , Orthomyxoviridae/drug effects , Silver/pharmacology , Tretinoin/pharmacology , Animals , Antiviral Agents/chemistry , Autophagy/drug effects , Cell Line, Tumor , Dogs , Epithelial Cells/metabolism , Epithelial Cells/virology , Humans , Lung/metabolism , Lung/virology , Madin Darby Canine Kidney Cells/drug effects , Madin Darby Canine Kidney Cells/virology , Microbial Sensitivity Tests , Mitochondria/metabolism , Silver/chemistry , Tretinoin/chemistry
11.
Am J Respir Cell Mol Biol ; 56(3): 372-382, 2017 03.
Article in English | MEDLINE | ID: mdl-27814452

ABSTRACT

The apical surface liquid (ASL) layer covers the airways and forms a first line of defense against pathogens. Maintenance of ASL volume by airway epithelia is essential for maintaining lung function. The proteolytic activation of epithelial Na+ channels is believed to be the dominating mechanism to cope with increases in ASL volumes. Alternative mechanisms, in particular increases in epithelial osmotic water permeability (Posm), have so far been regarded as rather less important. However, most studies mainly addressed immediate effects upon apical volume expansion (AVE) and increases in ASL. This study addresses the response of lung epithelia to long-term AVE. NCI-H441 cells and primary human tracheal epithelial cells, both cultivated in air-liquid interface conditions, were used as models for the lung epithelium. AVE was established by adding isotonic solution to the apical surface of differentiated lung epithelia, and time course of ASL volume restoration was assessed by the deuterium oxide dilution method. Concomitant ion transport was investigated in Ussing chambers. We identified a low resorptive state immediately after AVE, which coincided with proteolytic ion transport activation within 10-15 minutes after AVE. The main clearance of excess ASL occurred during a delayed (hours after AVE) high resorptive state, which did not correlate with ion transport activation. Instead, high resorptive state onset coincided with an increase in Posm, which depended on aquaporin up-regulation. In summary, our data demonstrate that, aside from ion transport activation, modulation of Posm is a major mechanism to compensate for long-term AVE in lung epithelia.


Subject(s)
Epithelium/metabolism , Lung/metabolism , Rheology , Water/metabolism , Amiloride/pharmacology , Aquaporins/metabolism , Epithelial Sodium Channels/metabolism , Epithelium/drug effects , Humans , Immunohistochemistry , Lung/drug effects , Osmosis/drug effects , Permeability/drug effects , Protease Inhibitors/pharmacology , Rheology/drug effects , Surface Properties , Time Factors
12.
Biochim Biophys Acta ; 1863(8): 2124-34, 2016 08.
Article in English | MEDLINE | ID: mdl-27155084

ABSTRACT

The molecular basis involving adsorption of pulmonary surfactant at the respiratory air-liquid interface and the specific roles of the surfactant proteins SP-B and SP-C in this process have not been completely resolved. The reasons might be found in the largely unknown structural assembly in which surfactant lipids and proteins are released from alveolar type II cells, and the difficulties to sample, manipulate and visualize the adsorption of these micron-sized particles at an air-liquid interface under appropriate physiological conditions. Here, we introduce several approaches to overcome these problems. First, by immunofluorescence we could demonstrate the presence of SP-B and SP-C on the surface of exocytosed surfactant particles. Second, by sampling the released particles and probing their adsorptive capacity we could demonstrate a remarkably high rate of interfacial adsorption, whose rate and extent was dramatically affected by treatment with antibodies against SP-B and SP-C. The effect of both antibodies was additive and specific. Third, direct microscopy of an inverted air-liquid interface revealed that the blocking effect is due to a stabilization of the released particles when contacting the air-liquid interface, precluding their transformation and the formation of surface films. We conclude that SP-B and SP-C are acting as essential, preformed molecular keys in the initial stages of surfactant unpacking and surface film formation. We further propose that surfactant activation might be transduced by a conformational change of the surfactant proteins upon contact with surface forces acting on the air-liquid interface.


Subject(s)
Alveolar Epithelial Cells/metabolism , Pulmonary Surfactant-Associated Protein B/physiology , Pulmonary Surfactant-Associated Protein C/physiology , Adsorption , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/ultrastructure , Animals , Boron Compounds , Cells, Cultured , Exocytosis , Fluorescent Dyes , Heterocyclic Compounds, 3-Ring , Hydrophobic and Hydrophilic Interactions , Microscopy, Confocal , Organelles/drug effects , Organelles/metabolism , Pulmonary Surfactant-Associated Protein B/antagonists & inhibitors , Pulmonary Surfactant-Associated Protein B/pharmacology , Pulmonary Surfactant-Associated Protein C/antagonists & inhibitors , Pulmonary Surfactant-Associated Protein C/pharmacology , Pulmonary Surfactants/chemistry , Rats , Rats, Sprague-Dawley , Surface Properties , Surface Tension
13.
Am J Respir Cell Mol Biol ; 54(5): 707-17, 2016 05.
Article in English | MEDLINE | ID: mdl-26473470

ABSTRACT

The lung epithelium constitutes a selective barrier that separates the airways from the aqueous interstitial compartment. Regulated barrier function controls water and ion transport across the epithelium and is essential for maintaining lung function. Tight junctions (TJs) seal the epithelial barrier and determine the paracellular transport. The properties of TJs depend especially on their claudin composition. Steroids are potent drugs used to treat a variety of airway diseases. Therefore, we addressed whether steroid hormones directly act on TJ properties in lung epithelia. Primary human tracheal epithelial cells and NCI-H441 cells, both cultivated under air-liquid interface conditions, were used as epithelial cell models. Our results demonstrate that glucocorticoids, but not mineralocorticoids, decreased paracellular permeability and shifted the ion permselectivity of TJs toward Cl(-). Glucocorticoids up-regulated claudin 8 (cldn8) expression via glucocorticoid receptors. Silencing experiments revealed that cldn8 is necessary to recruit occludin at the TJs. Immunohistochemistry on human lung tissue showed that cldn8 is specifically expressed in resorptive epithelia of the conducting and respiratory airways but not in the alveolar epithelium. We conclude that glucocorticoids enhance lung epithelia barrier function and increase paracellular Cl(-) selectivity via modulation of cldn8-dependent recruitment of occludin at the TJs. This mode of glucocorticoid action on lung epithelia might be beneficial to patients who suffer from impaired lung barrier function in various diseased conditions.


Subject(s)
Claudins/metabolism , Epithelium/metabolism , Glucocorticoids/pharmacology , Lung/metabolism , Tight Junctions/metabolism , Electric Impedance , Epithelium/drug effects , Fluorescent Antibody Technique , Gene Silencing/drug effects , Humans , Permeability/drug effects , RNA, Small Interfering/metabolism , Tight Junctions/drug effects , Time Factors , Up-Regulation/drug effects
14.
Cell Calcium ; 58(6): 628-37, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26560688

ABSTRACT

Ambroxol (Ax) is a frequently prescribed drug used to facilitate mucociliary clearance, but its mode of action is yet poorly understood. Here we show by X-ray spectroscopy that Ax accumulates in lamellar bodies (LBs), the surfactant storing, secretory lysosomes of type II pneumocytes. Using lyso- and acidotropic substances in combination with fluorescence imaging we confirm that these vesicles belong to the class of acidic Ca(2+) stores. Ax lead to a significant neutralization of LB pH, followed by intracellular Ca(2+) release, and to a dose-dependent surfactant exocytosis. Ax-induced Ca(2+) release was significantly reduced and slowed down by pretreatment of the cells with bafilomycin A1 (Baf A1), an inhibitor of the vesicular H(+) ATPase. These results could be nearly reproduced with NH3/NH4(+). The findings suggest that Ax accumulates within LBs and severely affects their H(+) and Ca(2+) homeostasis. This is further supported by an Ax-induced change of nanostructural assembly of surfactant layers. We conclude that Ax profoundly affects LBs presumably by disordering lipid bilayers and by acting as a weak base. The pH change triggers - at least in part - Ca(2+) release from stores and secretion of surfactant from type II cells. This novel mechanism of Ax as a lysosomal secretagogue may also play a role for its recently discussed use for lysosomal storage and other degenerative diseases.


Subject(s)
Ambroxol/pharmacology , Calcium/metabolism , Exocytosis/drug effects , Lysosomes/drug effects , Secretory Vesicles/drug effects , Alveolar Epithelial Cells , Animals , Cell Separation/methods , Exocytosis/physiology , Hydrogen-Ion Concentration , Rats, Sprague-Dawley
15.
Shock ; 43(2): 140-7, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25526372

ABSTRACT

Blunt chest trauma causes pulmonary and systemic inflammation. It is still a matter of debate whether the long-term course of this inflammatory response is associated with persistent impairment of lung function. We hypothesized that an increase of inflammatory biomarkers may still be present at later time points after blunt chest trauma, eventually, despite normalized lung mechanics and gas exchange. Anesthetized spontaneously breathing male C57BL/6J mice underwent a blast wave-induced blunt chest trauma or sham procedure. Twelve and 24 h later, blood gases and lung mechanics were measured, together with blood, bronchoalveolar lavage (BAL), and tissue cytokine concentrations (multiplex cytokine kit); heme oxygenase 1 (HO-1), activated caspase-3, Bcl-xL, and Bax expression (Western blotting); nuclear factor-κB activation (electrophoretic mobility shift assay); nitrotyrosine formation; and purinergic (P2XR4 and P2XR7) receptor expression (immunohistochemistry). Histological damage was assessed by hematoxylin and eosin and periodic acid-Schiff staining. High-resolution respirometry allowed assessing mitochondrial respiration in diaphragm biopsies. Chest trauma significantly increased tissue and BAL cytokine levels, associated with a significant increase in HO-1, purinergic receptor expression, and tissue nitrotyrosine formation. In contrast, lung mechanics, gas exchange, and histological damage did not show any significant difference between sham and trauma groups. Activation of the immune response remains present at later time points after murine blunt chest trauma. Discordance of the increased local inflammatory response and preserved pulmonary function may be explained by a dissociation of the immune response and lung function, such as previously suggested after experimental sepsis.


Subject(s)
Thoracic Injuries/physiopathology , Wounds, Nonpenetrating/physiopathology , Animals , Apoptosis/physiology , Cell Respiration/physiology , Chemokines/metabolism , Cytokines/metabolism , Disease Models, Animal , Lung/immunology , Lung/pathology , Lung/physiopathology , Male , Mice, Inbred C57BL , Mitochondria, Muscle/physiology , Pulmonary Gas Exchange/physiology , Receptors, Purinergic/biosynthesis , Respiratory Mechanics/physiology , Thoracic Injuries/immunology , Thoracic Injuries/pathology , Tyrosine/analogs & derivatives , Tyrosine/biosynthesis , Wounds, Nonpenetrating/immunology , Wounds, Nonpenetrating/pathology
16.
Crit Care ; 18(3): 149, 2014 Jun 10.
Article in English | MEDLINE | ID: mdl-25171553

ABSTRACT

It is well established that prolonged, controlled mechanical ventilation is associated with contractile dysfunction of the diaphragm due to impaired function of the mitochondrial respiratory chain as a result of aggravated oxidative and nitrosative stress. Sepsis and circulatory failure induce a similar response pattern. Callahan and Supinski now show that streptozotocin-induced insulin-dependent diabetes causes a comparable response pattern, both with respect to function and physiology - that is, reduced fiber force and, consequently, muscle contractility - but also as far as the underlying mechanisms are concerned. In other words, the authors elegantly demonstrate that the consequences of a chronic metabolic disease and that of acute critical illness may lead to the same phenotype response. It remains to be elucidated whether the underlying co-morbidity (for example, diabetes) adds to or even synergistically enhances the effect of an acute stress situation (for example, sepsis, mechanical ventilation). In addition, extending their previous work during shock states, the authors also show that administration of a preparation of the enzymatic anti-oxidant superoxide dismutase can reverse the deleterious effects of diabetes. These data are discussed in the context of the fundamental role of hyperglycemia in relation to metabolism-dependent formation of reactive oxygen species.


Subject(s)
Diaphragm/metabolism , Hyperglycemia/complications , Hyperglycemia/metabolism , Muscle Weakness/etiology , Muscle Weakness/metabolism , Oxidative Stress/physiology , Animals , Male
17.
Physiol Rep ; 2(1): e00201, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24744880

ABSTRACT

Proper apical airway surface hydration is essential to maintain lung function. This hydration depends on well-balanced water resorption and secretion. The mechanisms involved in resorption are still a matter of debate, especially as the measurement of transepithelial water transport remains challenging. In this study, we combined classical short circuit current (I SC) measurements with a novel D2O dilution method to correlate ion and water transport in order to reveal basic transport mechanisms in lung epithelia. D2O dilution method enabled precise analysis of water resorption with an unprecedented resolution. NCI-H441 cells cultured at an air-liquid interface resorbed water at a rate of 1.5 ± 0.4 µL/(h cm(2)). Water resorption and I SC were reduced by almost 80% in the presence of the bulk Cl(-) channel inhibitor 5-nitro-2-(3-phenylpropylamino)benzoic acid (NPPB) or amiloride, a specific inhibitor of epithelial sodium channel (ENaC). However, water resorption and I SC were only moderately affected by forskolin or cystic fibrosis transmembrane regulator (CFTR) channel inhibitors (CFTRinh-172 and glybenclamide). In line with previous studies, we demonstrate that water resorption depends on ENaC, and CFTR channels have only a minor but probably modulating effect on water resorption. However, the major ENaC-mediated water resorption depends on an apical non-CFTR Cl(-) conductance.

18.
Anal Chem ; 85(9): 4247-50, 2013 May 07.
Article in English | MEDLINE | ID: mdl-23560702

ABSTRACT

Lung epithelia regulate the water flux between gas filled airways and the interstitial compartment in order to maintain organ function. Current methodology to assess transepithelial water transport is limited. We present a D2O dilution method to quantify submicroliter volumes of aqueous solutions on epithelial cell layers. Evaluating D2O/H2O mixtures using mid-infrared (2-25 µm) attenuated total reflection (ATR) spectroscopy, with a resolution of 0.06% vol/vol change, corresponding to 24 nL, was achieved. Using this method, we demonstrate that water transport across NCI-H441 lung epithelial cell layers and apical surface liquid (ASL) volumes are coupled to dexamethasone dependent amiloride-sensitive ion transport. However, contrary to current dogma, electrogenic transport is not rate-limiting for water transport. This clearly indicates the need to directly assess net water rather than ion transport across epithelial cell layers. The presented D2O dilution method enables such direct and quick quantification of transepithelial water transport with high resolution.


Subject(s)
Adipose Tissue/metabolism , Deuterium Oxide , Epithelial Cells/metabolism , Lung/metabolism , Spectroscopy, Fourier Transform Infrared/methods , Water/metabolism , Biological Transport , Deuterium Oxide/analysis , Humans , Indicator Dilution Techniques
19.
FASEB J ; 27(4): 1772-83, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23307836

ABSTRACT

Two fundamental mechanisms within alveoli are essential for lung function: regulated fluid transport and secretion of surfactant. Surfactant is secreted via exocytosis of lamellar bodies (LBs) in alveolar type II (ATII) cells. We recently reported that LB exocytosis results in fusion-activated cation entry (FACE) via P2X4 receptors on LBs. We propose that FACE, in addition to facilitating surfactant secretion, modulates alveolar fluid transport. Correlative fluorescence and atomic force microscopy revealed that FACE-dependent water influx correlated with individual fusion events in rat primary ATII cells. Moreover, ATII cell monolayers grown at air-liquid interface exhibited increases in short-circuit current (Isc) on stimulation with ATP or UTP. Both are potent agonists for LB exocytosis, but only ATP activates FACE. ATP, not UTP, elicited additional fusion-dependent increases in Isc. Overexpressing dominant-negative P2X4 abrogated this effect by ∼50%, whereas potentiating P2X4 lead to ∼80% increase in Isc. Finally, we monitored changes in alveolar surface liquid (ASL) on ATII monolayers by confocal microscopy. Only stimulation with ATP, not UTP, led to a significant, fusion-dependent, 20% decrease in ASL, indicating apical-to-basolateral fluid transport across ATII monolayers. Our data support the first direct link between LB exocytosis, regulation of surfactant secretion, and transalveolar fluid resorption via FACE.


Subject(s)
Adenosine Triphosphate/pharmacology , Membrane Fusion/drug effects , Pulmonary Alveoli/metabolism , Pulmonary Surfactants/metabolism , Receptors, Purinergic P2X4/metabolism , Animals , Biological Transport/drug effects , Cations/metabolism , Exocytosis/physiology , Lung/drug effects , Lung/metabolism , Microscopy, Atomic Force/methods , Pulmonary Alveoli/cytology , Rats , Rats, Sprague-Dawley , Uridine Triphosphate/pharmacology
20.
FASEB J ; 27(4): 1322-9, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23241311

ABSTRACT

Keratin filaments impart resilience against mechanical extension of the cell. Despite the pathophysiological relevance of this function, very little is known about the mechanical properties of intermediate filaments in living cells and how these properties are modulated. We used keratin mutants that mimic or abrogate phosphorylation of keratin 8-serine(431) and keratin 18-serine(52) and investigated their effect on keratin tortuousness after cell stretch release in squamous cell carcinoma cells. Cells transfected with the wild-type keratins were used as controls. We can show that keratin dephosphorylation alters the stretch response of keratin in living cells since keratin tortuousness was abolished when phosphorylation of keratin18-serine(52) was abrogated. Additional experiments demonstrate that keratin tortuousness is not simply caused by a plastic overextension of keratin filaments because tortuousness is reversible and requires an intact actin-myosin system. The role of actin in this process remains unclear, but we suggest anchorage of keratin filaments to actin during stretch that leads to buckling on stretch release. Dephosphorylated keratin18-serine(52) might strengthen the recoil force of keratin filaments and hence explain the abolished buckling. The almost exclusive immunolabeling for phosphorylated keratin18-serine (52) in the cell periphery points at a particular role of the peripheral keratin network in this regard.


Subject(s)
Actins/metabolism , Epithelial Cells/metabolism , Intermediate Filaments/metabolism , Keratins/metabolism , Cell Survival , Cells, Cultured , Epithelial Cells/cytology , Humans , Keratins/ultrastructure , Phosphorylation/physiology , Serine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...