Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Sensors (Basel) ; 23(12)2023 Jun 07.
Article in English | MEDLINE | ID: mdl-37420569

ABSTRACT

In the paper, the effect of spontaneous Brillouin scattering (SpBS) is analyzed as a noise source in distributed acoustic sensors (DAS). The intensity of the SpBS wave fluctuates over time, and these fluctuations increase the noise power in DAS. Based on experimental data, the probability density function (PDF) of the spectrally selected SpBS Stokes wave intensity is negative exponential, which corresponds to the known theoretical conception. Based on this statement, an estimation of the average noise power induced by the SpBS wave is given. This noise power equals the square of the average power of the SpBS Stokes wave, which in turn is approximately 18 dB lower than the Rayleigh backscattering power. The noise composition in DAS is determined for two configurations, the first for the initial backscattering spectrum and the second for the spectrum in which the SpBS Stokes and anti-Stokes waves are rejected. It is established that in the analyzed particular case, the SpBS noise power is dominant and exceeds the powers of the thermal, shot, and phase noises in DAS. Accordingly, by rejecting the SpBS waves at the photodetector input, it is possible to reduce the noise power in DAS. In our case, this rejection is carried out by an asymmetric Mach-Zehnder interferometer (MZI). The rejection of the SpBS wave is most relevant for broadband photodetectors, which are associated with the use of short probing pulses to achieve short gauge lengths in DAS.


Subject(s)
Fertilization , Heart Rate , Likelihood Functions
2.
Biomedicines ; 11(6)2023 Jun 05.
Article in English | MEDLINE | ID: mdl-37371733

ABSTRACT

Vascular barrier dysfunction is characterized by increased permeability and inflammation of endothelial cells (ECs), which are prominent features of acute lung injury (ALI), acute respiratory distress syndrome (ARDS), and sepsis, and a major complication of the SARS-CoV-2 infection and COVID-19. Functional impairment of the EC barrier and accompanying inflammation arises due to microbial toxins and from white blood cells of the lung as part of a defensive action against pathogens, ischemia-reperfusion or blood product transfusions, and aspiration syndromes-based injury. A loss of barrier function results in the excessive movement of fluid and macromolecules from the vasculature into the interstitium and alveolae resulting in pulmonary edema and collapse of the architecture and function of the lungs, and eventually culminates in respiratory failure. Therefore, EC barrier integrity, which is heavily dependent on cytoskeletal elements (mainly actin filaments, microtubules (MTs), cell-matrix focal adhesions, and intercellular junctions) to maintain cellular contacts, is a critical requirement for the preservation of lung function. EC cytoskeletal remodeling is regulated, at least in part, by Ser/Thr phosphorylation/dephosphorylation of key cytoskeletal proteins. While a large body of literature describes the role of phosphorylation of cytoskeletal proteins on Ser/Thr residues in the context of EC barrier regulation, the role of Ser/Thr dephosphorylation catalyzed by Ser/Thr protein phosphatases (PPases) in EC barrier regulation is less documented. Ser/Thr PPases have been proposed to act as a counter-regulatory mechanism that preserves the EC barrier and opposes EC contraction. Despite the importance of PPases, our knowledge of the catalytic and regulatory subunits involved, as well as their cellular targets, is limited and under-appreciated. Therefore, the goal of this review is to discuss the role of Ser/Thr PPases in the regulation of lung EC cytoskeleton and permeability with special emphasis on the role of protein phosphatase 1 (PP1) and protein phosphatase 2A (PP2A) as major mammalian Ser/Thr PPases. Importantly, we integrate the role of PPases with the structural dynamics of the cytoskeleton and signaling cascades that regulate endothelial cell permeability and inflammation.

3.
Sensors (Basel) ; 22(23)2022 Dec 04.
Article in English | MEDLINE | ID: mdl-36502184

ABSTRACT

A simple and cost-effective architecture of a distributed acoustic sensor (DAS) or a phase-OTDR for engineering geology is proposed. The architecture is based on the dual-pulse acquisition principle, where the dual probing pulse is formed via an unbalanced Michelson interferometer (MI). The necessary phase shifts between the sub-pulses of the dual-pulse are introduced using a 3 × 3 coupler built into the MI. Laser pulses are generated by direct modulation of the injection current, which obtains optical pulses with a duration of 7 ns. The use of an unbalanced MI for the formation of a dual-pulse reduces the requirements for the coherence of the laser source, as the introduced delay between sub-pulses is compensated in the fiber under test (FUT). Therefore, a laser with a relatively broad spectral linewidth of about 1 GHz can be used. To overcome the fading problem, as well as to ensure the linearity of the DAS response, the averaging of over 16 optical frequencies is used. The performance of the DAS was tested by recording a strong vibration impact on a horizontally buried cable and by the recording of seismic waves in a borehole in the seabed.


Subject(s)
Engineering , Geology , Cost-Benefit Analysis , Heart Rate , Acoustics
4.
Sensors (Basel) ; 22(3)2022 Jan 28.
Article in English | MEDLINE | ID: mdl-35161779

ABSTRACT

This work presents a detailed review of the development of distributed acoustic sensors (DAS) and their newest scientific applications. It covers most areas of human activities, such as the engineering, material, and humanitarian sciences, geophysics, culture, biology, and applied mechanics. It also provides the theoretical basis for most well-known DAS techniques and unveils the features that characterize each particular group of applications. After providing a summary of research achievements, the paper develops an initial perspective of the future work and determines the most promising DAS technologies that should be improved.


Subject(s)
Acoustics , Fiber Optic Technology , Humans
5.
Anal Methods ; 13(21): 2424-2433, 2021 06 07.
Article in English | MEDLINE | ID: mdl-33998615

ABSTRACT

Hepatitis B surface antigen (HBsAg) is the most clinically relevant serological marker of hepatitis B virus (HBV) infection. Its detection in blood is extremely important for identification of asymptomatic individuals or chronic HBV carriers, screening blood donors, and early seroconversion. Rapid point-of-care HBsAg tests are predominantly qualitative, and their analytical sensitivity does not meet the requirements of regulatory agencies. We present a highly sensitive lateral flow assay based on superparamagnetic nanoparticles for rapid quantification (within 30 min) of polyvalent HBsAg in serum. The demonstrated limit of detection (LOD) of 80 pg mL-1 in human serum is better than both the FDA recommendations for HBsAg assays (which is 0.5 ng mL-1) and the sensitivity of traditional laboratory-based methods such as enzyme linked immunosorbent assays. Along with the attractive LOD at lower concentrations and the wide linear dynamic range of more than 2.5 orders, the assay features rapidity, user-friendliness, on-site operation and effective performance in the complex biological medium. These are due to the combination of the immunochromatographic approach with a highly sensitive electronic registration of superparamagnetic nanolabels over the entire volume of a 3D test structure by their non-linear magnetization and selection of optimal antibodies by original optical label-free methods. The developed cost-efficient bioanalytical technology can be used in many socially important fields such as out-of-lab screening and diagnosis of HBV infection at a point-of-demand, especially in hard-to-reach or sparsely populated areas, as well as highly endemic regions.


Subject(s)
Hepatitis B Surface Antigens , Hepatitis B virus , Hepatitis B Antibodies , Hepatitis B virus/genetics , Humans , Magnetic Iron Oxide Nanoparticles , Sensitivity and Specificity
6.
Toxins (Basel) ; 12(4)2020 04 02.
Article in English | MEDLINE | ID: mdl-32252376

ABSTRACT

Bacterial toxins play a key role in the pathogenesis of lung disease. Based on their structural and functional properties, they employ various strategies to modulate lung barrier function and to impair host defense in order to promote infection. Although in general, these toxins target common cellular signaling pathways and host compartments, toxin- and cell-specific effects have also been reported. Toxins can affect resident pulmonary cells involved in alveolar fluid clearance (AFC) and barrier function through impairing vectorial Na+ transport and through cytoskeletal collapse, as such, destroying cell-cell adhesions. The resulting loss of alveolar-capillary barrier integrity and fluid clearance capacity will induce capillary leak and foster edema formation, which will in turn impair gas exchange and endanger the survival of the host. Toxins modulate or neutralize protective host cell mechanisms of both the innate and adaptive immunity response during chronic infection. In particular, toxins can either recruit or kill central players of the lung's innate immune responses to pathogenic attacks, i.e., alveolar macrophages (AMs) and neutrophils. Pulmonary disorders resulting from these toxin actions include, e.g., acute lung injury (ALI), the acute respiratory syndrome (ARDS), and severe pneumonia. When acute infection converts to persistence, i.e., colonization and chronic infection, lung diseases, such as bronchitis, chronic obstructive pulmonary disease (COPD), and cystic fibrosis (CF) can arise. The aim of this review is to discuss the impact of bacterial toxins in the lungs and the resulting outcomes for pathogenesis, their roles in promoting bacterial dissemination, and bacterial survival in disease progression.


Subject(s)
Bacteria/pathogenicity , Bacterial Infections/microbiology , Bacterial Toxins/metabolism , Lung/microbiology , Respiratory Tract Infections/microbiology , Adaptive Immunity , Animals , Bacteria/immunology , Bacteria/metabolism , Bacterial Infections/immunology , Bacterial Infections/metabolism , Bacterial Infections/pathology , Disease Progression , Host-Pathogen Interactions , Humans , Immunity, Innate , Lung/immunology , Lung/metabolism , Lung/pathology , Respiratory Tract Infections/immunology , Respiratory Tract Infections/metabolism , Respiratory Tract Infections/pathology , Signal Transduction
7.
Front Immunol ; 9: 1309, 2018.
Article in English | MEDLINE | ID: mdl-29951058

ABSTRACT

Pneumonia is a leading cause of death in children and the elderly worldwide, accounting for 15% of all deaths of children under 5 years old. Streptococcus pneumoniae is a common and aggressive cause of pneumonia and can also contribute to meningitis and sepsis. Despite the widespread use of antibiotics, mortality rates for pneumonia remain unacceptably high in part due to the release of bacterial toxins. Pneumolysin (PLY) is a cholesterol-dependent toxin that is produced by Streptococcus, and it is both necessary and sufficient for the development of the extensive pulmonary permeability edema that underlies acute lung injury. The mechanisms by which PLY disrupts the pulmonary endothelial barrier are not fully understood. Previously, we found that reactive oxygen species (ROS) contribute to the barrier destructive effects of PLY and identified an unexpected but potent role of Hsp70 in suppressing ROS production. The ability of Hsp70 to influence PLY-induced barrier dysfunction is not yet described, and the goal of the current study was to identify whether Hsp70 upregulation is an effective strategy to protect the lung microvascular endothelial barrier from G+ bacterial toxins. Overexpression of Hsp70 via adenovirus-mediated gene transfer attenuated PLY-induced increases in permeability in human lung microvascular endothelial cells (HLMVEC) with no evidence of cytotoxicity. To adopt a more translational approach, we employed a pharmacological approach using geranylgeranylacetone (GGA) to acutely upregulate endogenous Hsp70 expression. Following acute treatment (6 h) with GGA, HLMVECs exposed to PLY displayed improved cell viability and enhanced endothelial barrier function as measured by both Electric Cell-substrate Impedance Sensing (ECIS) and transwell permeability assays compared to control treated cells. PLY promoted increased mitochondrial ROS, decreased mitochondrial oxygen consumption, and increased caspase 3 cleavage and cell death, which were collectively improved in cells pretreated with GGA. In mice, IP pretreatment with GGA 24 h prior to IT administration of PLY resulted in significantly less Evans Blue Dye extravasation compared to vehicle, indicating preserved endothelial barrier integrity and suggesting that the acute upregulation of Hsp70 may be an effective therapeutic approach in the treatment of lung injury associated with pneumonia.

8.
Toxins (Basel) ; 10(2)2018 02 11.
Article in English | MEDLINE | ID: mdl-29439494

ABSTRACT

Pulmonary permeability edema is characterized by reduced alveolar Na⁺ uptake capacity and capillary barrier dysfunction and is a potentially lethal complication of listeriosis. Apical Na⁺ uptake is mainly mediated by the epithelial sodium channel (ENaC) and initiates alveolar liquid clearance. Here we examine how listeriolysin O (LLO), the pore-forming toxin of Listeria monocytogenes, impairs the expression and activity of ENaC. To that purpose, we studied how sub-lytic concentrations of LLO affect negative and positive regulators of ENaC expression in the H441 airway epithelial cell line. LLO reduced expression of the crucial ENaC-α subunit in H441 cells within 2 h and this was preceded by activation of PKC-α, a negative regulator of the channel's expression. At later time points, LLO caused a significant reduction in the phosphorylation of Sgk-1 at residue T256 and of Akt-1 at residue S473, both of which are required for full activation of ENaC. The TNF-derived TIP peptide prevented LLO-mediated PKC-α activation and restored phospho-Sgk-1-T256. The TIP peptide also counteracted the observed LLO-induced decrease in amiloride-sensitive Na⁺ current and ENaC-α expression in H441 cells. Intratracheally instilled LLO caused profound pulmonary edema formation in mice, an effect that was prevented by the TIP peptide; thus indicating the therapeutic potential of the peptide for the treatment of pore-forming toxin-associated permeability edema.


Subject(s)
Bacterial Toxins/toxicity , Epithelial Sodium Channels/physiology , Heat-Shock Proteins/toxicity , Hemolysin Proteins/toxicity , Peptides/pharmacology , Peptides/therapeutic use , Pulmonary Edema/drug therapy , Animals , Bronchi/cytology , Cell Line , Epithelial Cells/drug effects , Epithelial Cells/physiology , Humans , Immediate-Early Proteins/metabolism , Male , Mice, Inbred C57BL , Phosphorylation/drug effects , Protein Kinase C-alpha/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism
9.
Am J Respir Cell Mol Biol ; 58(5): 614-624, 2018 05.
Article in English | MEDLINE | ID: mdl-29115856

ABSTRACT

One of the early events in the progression of LPS-mediated acute lung injury in mice is the disruption of the pulmonary endothelial barrier resulting in lung edema. However, the molecular mechanisms by which the endothelial barrier becomes compromised remain unresolved. The SRY (sex-determining region on the Y chromosome)-related high-mobility group box (Sox) group F family member, SOX18, is a barrier-protective protein through its ability to increase the expression of the tight junction protein CLDN5. Thus, the purpose of this study was to determine if downregulation of the SOX18-CLDN5 axis plays a role in the pulmonary endothelial barrier disruption associated with LPS exposure. Our data indicate that both SOX18 and CLDN5 expression is decreased in two models of in vivo LPS exposure (intraperitoneal, intratracheal). A similar downregulation was observed in cultured human lung microvascular endothelial cells (HLMVECs) exposed to LPS. SOX18 overexpression in HLMVECs or in the mouse lung attenuated the LPS-mediated vascular barrier disruption. Conversely, reduced CLDN5 expression (siRNA) reduced the HLMVEC barrier-protective effects of SOX18 overexpression. The mechanism by which LPS decreases SOX18 expression was identified as transcriptional repression through binding of NF-κB (p65) to a SOX18 promoter sequence located between -1,082 and -1,073 bp with peroxynitrite contributing to LPS-mediated NF-κB activation. We conclude that NF-κB-dependent decreases in the SOX18-CLDN5 axis are essentially involved in the disruption of human endothelial cell barrier integrity associated with LPS-mediated acute lung injury.


Subject(s)
Acute Lung Injury/metabolism , Capillary Permeability , Endothelial Cells/metabolism , Lipopolysaccharides , Lung/blood supply , NF-kappa B/metabolism , Pulmonary Edema/metabolism , SOXF Transcription Factors/metabolism , Acute Lung Injury/chemically induced , Acute Lung Injury/genetics , Acute Lung Injury/pathology , Animals , Binding Sites , Cells, Cultured , Claudin-5/genetics , Claudin-5/metabolism , Disease Models, Animal , Down-Regulation , Endothelial Cells/pathology , Humans , Male , Mice, Inbred C57BL , NF-kappa B/genetics , Peroxynitrous Acid/metabolism , Promoter Regions, Genetic , Protein Binding , Pulmonary Edema/chemically induced , Pulmonary Edema/genetics , Pulmonary Edema/pathology , SOXF Transcription Factors/genetics , Signal Transduction , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism
10.
Front Immunol ; 8: 842, 2017.
Article in English | MEDLINE | ID: mdl-28785264

ABSTRACT

BACKGROUND: Streptococcus pneumoniae is a major etiologic agent of bacterial pneumonia. Autolysis and antibiotic-mediated lysis of pneumococci induce release of the pore-forming toxin, pneumolysin (PLY), their major virulence factor, which is a prominent cause of acute lung injury. PLY inhibits alveolar liquid clearance and severely compromises alveolar-capillary barrier function, leading to permeability edema associated with pneumonia. As a consequence, alveolar flooding occurs, which can precipitate lethal hypoxemia by impairing gas exchange. The α subunit of the epithelial sodium channel (ENaC) is crucial for promoting Na+ reabsorption across Na+-transporting epithelia. However, it is not known if human lung microvascular endothelial cells (HL-MVEC) also express ENaC-α and whether this subunit is involved in the regulation of their barrier function. METHODS: The presence of α, ß, and γ subunits of ENaC and protein phosphorylation status in HL-MVEC were assessed in western blotting. The role of ENaC-α in monolayer resistance of HL-MVEC was examined by depletion of this subunit by specific siRNA and by employing the TNF-derived TIP peptide, a specific activator that directly binds to ENaC-α. RESULTS: HL-MVEC express all three subunits of ENaC, as well as acid-sensing ion channel 1a (ASIC1a), which has the capacity to form hybrid non-selective cation channels with ENaC-α. Both TIP peptide, which specifically binds to ENaC-α, and the specific ASIC1a activator MitTx significantly strengthened barrier function in PLY-treated HL-MVEC. ENaC-α depletion significantly increased sensitivity to PLY-induced hyperpermeability and in addition, blunted the protective effect of both the TIP peptide and MitTx, indicating an important role for ENaC-α and for hybrid NSC channels in barrier function of HL-MVEC. TIP peptide blunted PLY-induced phosphorylation of both calmodulin-dependent kinase II (CaMKII) and of its substrate, the actin-binding protein filamin A (FLN-A), requiring the expression of both ENaC-α and ASIC1a. Since non-phosphorylated FLN-A promotes ENaC channel open probability and blunts stress fiber formation, modulation of this activity represents an attractive target for the protective actions of ENaC-α in both barrier function and liquid clearance. CONCLUSION: Our results in cultured endothelial cells demonstrate a previously unrecognized role for ENaC-α in strengthening capillary barrier function that may apply to the human lung. Strategies aiming to activate endothelial NSC channels that contain ENaC-α should be further investigated as a novel approach to improve barrier function in the capillary endothelium during pneumonia.

11.
Sci Rep ; 6: 39018, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27976727

ABSTRACT

The goal of this study was to investigate the role of MLC phosphatase (MLCP) in a LPS model of acute lung injury (ALI). We demonstrate that ectopic expression of a constitutively-active (C/A) MLCP regulatory subunit (MYPT1) attenuates the ability of LPS to increase endothelial (EC) permeability. Down-regulation of MYPT1 exacerbates LPS-induced expression of ICAM1 suggesting an anti-inflammatory role of MLCP. To determine whether MLCP contributes to LPS-induced ALI in vivo, we utilized a nanoparticle DNA delivery method to specifically target lung EC. Expression of a C/A MYPT1 reduced LPS-induced lung inflammation and vascular permeability. Further, increased expression of the CS1ß (MLCP catalytic subunit) also reduced LPS-induced lung inflammation, whereas the inactive CS1ß mutant increased vascular leak. We next examined the role of the cytoskeletal targets of MLCP, the ERM proteins (Ezrin/Radixin/Moesin), in mediating barrier dysfunction. LPS-induced increase in EC permeability was accompanied by PKC-mediated increase in ERM phosphorylation, which was more prominent in CS1ß-depleted cells. Depletion of Moesin and Ezrin, but not Radixin attenuated LPS-induced increases in permeability. Further, delivery of a Moesin phospho-null mutant into murine lung endothelium attenuated LPS-induced lung inflammation and vascular leak suggesting that MLCP opposes LPS-induced ALI by mediating the dephosphorylation of Moesin and Ezrin.


Subject(s)
Acute Lung Injury , Cytoskeletal Proteins/metabolism , Endothelium, Vascular/metabolism , Lipopolysaccharides/toxicity , Membrane Proteins/metabolism , Microfilament Proteins/metabolism , Myosin-Light-Chain Phosphatase/metabolism , Acute Lung Injury/chemically induced , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Acute Lung Injury/prevention & control , Animals , Capillary Permeability/drug effects , Endothelium, Vascular/pathology , Humans , Mice , Phosphorylation
12.
Anal Bioanal Chem ; 407(14): 3955-64, 2015 May.
Article in English | MEDLINE | ID: mdl-25757827

ABSTRACT

A 3-channel biosensor based on spectral correlation interferometry (SCI) has been adapted for direct optical detection of antigens by measuring changes in thickness of a biolayer on functionalized glass slips employed as affordable single-use sensor chips. The instrument is insensitive to the bulk refractive index of a solution under test and provides signals in metrological units (pm or nm). Using real-time monitoring with the SCI, protocols for fabrication of sensor chips with different functional (epoxylated, carboxylated, and biotinylated) surfaces for antibody immobilization have been developed and optimized to minimize chip-to-chip variations and achieve better limit of detection (LOD), shorter assay time, and longer shelf life. The optimized coupling surfaces have been compared for detection of human serum albumin (HSA) used as a model agent of medical significance. The dynamic ranges for measuring the HSA concentration were 0.07-20, 0.12-30, and 0.25-10 µg/ml, and the assay durations were less than 20, 15, and 30 min for the epoxylated, carboxylated, and biotinylated chips, respectively. The advantages of each type of sensor chip have been shown, namely, the carboxylated chips feature the shortest assay time, the epoxylated ones demonstrate the best LOD, and the biotinylated chips exhibit the longest shelf life in an unprotected environment. The developed protocols of antibody immobilization can be used in different biosensors and assay techniques including those based on fluorescent, magnetic or plasmonic labels, etc. The SCI is well compatible with various partially transparent layers used in biosensing and with microarrays for multi-analyte detection.


Subject(s)
Antibodies, Immobilized , Biosensing Techniques/instrumentation , Biosensing Techniques/methods , Interferometry/methods , Serum Albumin/chemistry , Humans , Interferometry/instrumentation , Lab-On-A-Chip Devices , Limit of Detection
13.
Front Physiol ; 5: 259, 2014.
Article in English | MEDLINE | ID: mdl-25076911

ABSTRACT

RATIONALE: Antibiotic treatment of patients infected with G(-) or G(+) bacteria promotes release of the toxins lipopolysaccharide (LPS) and pneumolysin (PLY) in their lungs. Growth Hormone-releasing Hormone (GHRH) agonist JI-34 protects human lung microvascular endothelial cells (HL-MVEC), expressing splice variant 1 (SV-1) of the receptor, from PLY-induced barrier dysfunction. We investigated whether JI-34 also blunts LPS-induced hyperpermeability. Since GHRH receptor (GHRH-R) signaling can potentially stimulate both cAMP-dependent barrier-protective pathways as well as barrier-disruptive protein kinase C pathways, we studied their interaction in GHRH agonist-treated HL-MVEC, in the presence of PLY, by means of siRNA-mediated protein kinase A (PKA) depletion. METHODS: Barrier function measurements were done in HL-MVEC monolayers using Electrical Cell substrate Impedance Sensing (ECIS) and VE-cadherin expression by Western blotting. Capillary leak was assessed by Evans Blue dye (EBD) incorporation. Cytokine generation in broncho-alveolar lavage fluid (BALF) was measured by multiplex analysis. PKA and PKC-α activity were assessed by Western blotting. RESULTS: GHRH agonist JI-34 significantly blunts LPS-induced barrier dysfunction, at least in part by preserving VE-cadherin expression, while not affecting inflammation. In addition to activating PKA, GHRH agonist also increases PKC-α activity in PLY-treated HL-MVEC. Treatment with PLY significantly decreases resistance in control siRNA-treated HL-MVEC, but does so even more in PKA-depleted monolayers. Pretreatment with GHRH agonist blunts PLY-induced permeability in control siRNA-treated HL-MVEC, but fails to improve barrier function in PKA-depleted PLY-treated monolayers. CONCLUSIONS: GHRH signaling in HL-MVEC protects from both LPS and PLY-mediated endothelial barrier dysfunction and concurrently induces a barrier-protective PKA-mediated and a barrier-disruptive PKC-α-induced pathway in the presence of PLY, the former of which dominates the latter.

14.
Am J Respir Crit Care Med ; 190(5): 522-32, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-25029038

ABSTRACT

RATIONALE: Alveolar liquid clearance is regulated by Na(+) uptake through the apically expressed epithelial sodium channel (ENaC) and basolaterally localized Na(+)-K(+)-ATPase in type II alveolar epithelial cells. Dysfunction of these Na(+) transporters during pulmonary inflammation can contribute to pulmonary edema. OBJECTIVES: In this study, we sought to determine the precise mechanism by which the TIP peptide, mimicking the lectin-like domain of tumor necrosis factor (TNF), stimulates Na(+) uptake in a homologous cell system in the presence or absence of the bacterial toxin pneumolysin (PLY). METHODS: We used a combined biochemical, electrophysiological, and molecular biological in vitro approach and assessed the physiological relevance of the lectin-like domain of TNF in alveolar liquid clearance in vivo by generating triple-mutant TNF knock-in mice that express a mutant TNF with deficient Na(+) uptake stimulatory activity. MEASUREMENTS AND MAIN RESULTS: TIP peptide directly activates ENaC, but not the Na(+)-K(+)-ATPase, upon binding to the carboxy-terminal domain of the α subunit of the channel. In the presence of PLY, a mediator of pneumococcal-induced pulmonary edema, this binding stabilizes the ENaC-PIP2-MARCKS complex, which is necessary for the open probability conformation of the channel and preserves ENaC-α protein expression, by means of blunting the protein kinase C-α pathway. Triple-mutant TNF knock-in mice are more prone than wild-type mice to develop edema with low-dose intratracheal PLY, correlating with reduced pulmonary ENaC-α subunit expression. CONCLUSIONS: These results demonstrate a novel TNF-mediated mechanism of direct ENaC activation and indicate a physiological role for the lectin-like domain of TNF in the resolution of alveolar edema during inflammation.


Subject(s)
Epithelial Sodium Channel Agonists/metabolism , Epithelial Sodium Channels/metabolism , Peptides, Cyclic/metabolism , Pulmonary Alveoli/metabolism , Pulmonary Edema/metabolism , Streptolysins , Tumor Necrosis Factor-alpha/metabolism , Animals , Bacterial Proteins , Epithelial Sodium Channel Agonists/chemistry , Epithelial Sodium Channels/chemistry , Female , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Peptides, Cyclic/chemistry , Pulmonary Alveoli/microbiology , Pulmonary Edema/microbiology , Tumor Necrosis Factor-alpha/chemistry
15.
Am J Physiol Lung Cell Mol Physiol ; 306(6): L497-507, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24414256

ABSTRACT

Acute lung injury and acute respiratory distress syndrome (ALI/ARDS) affect 200,000 people a year in the USA. Pulmonary vascular and specifically endothelial cell (EC) barrier compromise is a hallmark of these diseases. We have recently shown that extracellular adenosine enhances human pulmonary (EC) barrier via activation of adenosine receptors (ARs) in cell cultures. On the basis of these data, we hypothesized that activation of ARs might exert barrier-protective effects in a model of ALI/ARDS in mice. To test this hypothesis, we examined the effects of pre- and posttreatment of adenosine and 5'-N-ethylcarboxamidoadenosine (NECA), a nonselective stable AR agonist, on LPS-induced lung injury. Mice were given vehicle or LPS intratracheally followed by adenosine, NECA, or vehicle instilled via the internal jugular vein. Postexperiment cell counts, Evans Blue Dye albumin (EBDA) extravasation, levels of proteins, and inflammatory cytokines were analyzed. Harvested lungs were used for histology and myeloperoxidase studies. Mice challenged with LPS alone demonstrated an inflammatory response typical of ALI. Cell counts, EBDA extravasation, as well as levels of proteins and inflammatory cytokines were decreased in adenosine-treated mice. Histology displayed reduced infiltration of neutrophils. NECA had a similar effect on LPS-induced vascular barrier compromise. Importantly, posttreatment with adenosine or NECA recovers lung vascular barrier and reduces inflammation induced by LPS challenge. Furthermore, adenosine significantly attenuated protein degradation of A2A and A3 receptors induced by LPS. Collectively, our results demonstrate that activation of ARs protects and restores vascular barrier functions and reduces inflammation in LPS-induced ALI.


Subject(s)
Acute Lung Injury/metabolism , Adenosine/metabolism , Endothelium/metabolism , Receptors, Purinergic P1/metabolism , Acute Lung Injury/chemically induced , Adenosine-5'-(N-ethylcarboxamide)/metabolism , Animals , Bronchoalveolar Lavage Fluid/cytology , Capillary Permeability/drug effects , Cell Count , Cytokines/metabolism , Endothelial Cells/metabolism , Inflammation/drug therapy , Inflammation/metabolism , Interleukin-6/metabolism , Lipopolysaccharides , Lung/metabolism , Lung/physiology , Mice , Mice, Inbred C57BL , Purinergic P1 Receptor Agonists/metabolism , Respiratory Distress Syndrome/metabolism , Tumor Necrosis Factor-alpha/metabolism
16.
Am J Respir Cell Mol Biol ; 50(1): 170-9, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23972231

ABSTRACT

Permeability of the endothelial monolayer is increased when exposed to the bacterial endotoxin LPS. Our previous studies have shown that heat shock protein (Hsp) 90 inhibitors protect and restore LPS-mediated hyperpermeability in bovine pulmonary arterial endothelial cells. In this study, we assessed the effect of Hsp90 inhibition against LPS-mediated hyperpermeability in cultured human lung microvascular endothelial cells (HLMVECs) and delineated the underlying molecular mechanisms. We demonstrate that Hsp90 inhibition is critical in the early phase, to prevent LPS-mediated hyperpermeability, and also in the later phase, to restore LPS-mediated hyperpermeability in HLMVECs. Because RhoA is a well known mediator of endothelial hyperpermeability, we investigated the effect of Hsp90 inhibition on LPS-mediated RhoA signaling. RhoA nitration and activity were increased by LPS in HLMVECs and suppressed when pretreated with the Hsp90 inhibitor, 17-allylamino-17 demethoxy-geldanamycin (17-AAG). In addition, inhibition of Rho kinase, a downstream effector of RhoA, protected HLMVECs from LPS-mediated hyperpermeability and abolished LPS-induced myosin light chain (MLC) phosphorylation, a target of Rho kinase. In agreement with these findings, 17-AAG or dominant-negative RhoA attenuated LPS-induced MLC phosphorylation. MLC phosphorylation induced by constitutively active RhoA was also suppressed by 17-AAG, suggesting a role for Hsp90 downstream of RhoA. Inhibition of Src family kinases also suppressed RhoA activity and MLC phosphorylation. Together, these data indicate that Hsp90 inhibition prevents and repairs LPS-induced lung endothelial barrier dysfunction by suppressing Src-mediated RhoA activity and signaling.


Subject(s)
Endothelial Cells/drug effects , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/metabolism , Lipopolysaccharides/adverse effects , rho-Associated Kinases/metabolism , Animals , Benzoquinones/pharmacology , Capillary Permeability/drug effects , Cells, Cultured , Endothelial Cells/metabolism , Humans , Lactams, Macrocyclic/pharmacology , Male , Mice , Mice, Inbred C57BL , Myosin Light Chains/metabolism , Phosphorylation/drug effects , Signal Transduction/drug effects , rhoA GTP-Binding Protein/metabolism , src-Family Kinases/metabolism
17.
Am J Respir Cell Mol Biol ; 50(3): 614-25, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24134589

ABSTRACT

Acute lung injury (ALI) is a severe hypoxemic respiratory insufficiency associated with lung leak, diffuse alveolar damage, inflammation, and loss of lung function. Decreased dimethylaminohydrolase (DDAH) activity and increases in asymmetric dimethylarginine (ADMA), together with exaggerated oxidative/nitrative stress, contributes to the development of ALI in mice exposed to LPS. Whether restoring DDAH function and suppressing ADMA levels can effectively ameliorate vascular hyperpermeability and lung injury in ALI is unknown, and was the focus of this study. In human lung microvascular endothelial cells, DDAH II overexpression prevented the LPS-dependent increase in ADMA, superoxide, peroxynitrite, and protein nitration. DDAH II also attenuated the endothelial barrier disruption associated with LPS exposure. Similarly, in vivo, we demonstrated that the targeted overexpression of DDAH II in the pulmonary vasculature significantly inhibited the accumulation of ADMA and the subsequent increase in oxidative/nitrative stress in the lungs of mice exposed to LPS. In addition, augmenting pulmonary DDAH II activity before LPS exposure reduced lung vascular leak and lung injury and restored lung function when DDAH activity was increased after injury. Together, these data suggest that enhancing DDAH II activity may prove a useful adjuvant therapy to treat patients with ALI.


Subject(s)
Acute Lung Injury/prevention & control , Amidohydrolases/metabolism , Endothelial Cells/enzymology , Genetic Therapy , Lipopolysaccharides , Lung/blood supply , Microvessels/enzymology , Pulmonary Edema/prevention & control , Acute Lung Injury/chemically induced , Acute Lung Injury/enzymology , Acute Lung Injury/genetics , Amidohydrolases/genetics , Animals , Arginine/analogs & derivatives , Arginine/metabolism , Bronchoalveolar Lavage Fluid/chemistry , Capillary Permeability , Cells, Cultured , Disease Models, Animal , Endothelial Cells/pathology , Humans , Lung/enzymology , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Microvessels/pathology , Oxidative Stress , Peroxynitrous Acid/metabolism , Pulmonary Edema/chemically induced , Pulmonary Edema/enzymology , Pulmonary Edema/genetics , Superoxides/metabolism , Time Factors , Transfection , Up-Regulation
18.
Toxins (Basel) ; 5(7): 1244-60, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23860351

ABSTRACT

Severe pneumonia is the main single cause of death worldwide in children under five years of age. The main etiological agent of pneumonia is the G+ bacterium Streptococcus pneumoniae, which accounts for up to 45% of all cases. Intriguingly, patients can still die days after commencing antibiotic treatment due to the development of permeability edema, although the pathogen was successfully cleared from their lungs. This condition is characterized by a dramatically impaired alveolar epithelial-capillary barrier function and a dysfunction of the sodium transporters required for edema reabsorption, including the apically expressed epithelial sodium channel (ENaC) and the basolaterally expressed sodium potassium pump (Na+-K+-ATPase). The main agent inducing this edema formation is the virulence factor pneumolysin, a cholesterol-binding pore-forming toxin, released in the alveolar compartment of the lungs when pneumococci are being lysed by antibiotic treatment or upon autolysis. Sub-lytic concentrations of pneumolysin can cause endothelial barrier dysfunction and can impair ENaC-mediated sodium uptake in type II alveolar epithelial cells. These events significantly contribute to the formation of permeability edema, for which currently no standard therapy is available. This review focuses on discussing some recent developments in the search for the novel therapeutic agents able to improve lung function despite the presence of pore-forming toxins. Such treatments could reduce the potentially lethal complications occurring after antibiotic treatment of patients with severe pneumonia.


Subject(s)
Lung/microbiology , Pneumonia/therapy , Streptococcus pneumoniae/pathogenicity , Streptolysins/toxicity , Animals , Bacterial Proteins/toxicity , Child, Preschool , Disease Models, Animal , Growth Hormone/metabolism , Humans , Immune System/microbiology , Lectins/therapeutic use , Lung/pathology , Pneumonia/microbiology , Pulmonary Edema/microbiology , Pulmonary Edema/therapy , Structure-Activity Relationship , Tumor Necrosis Factor-alpha/metabolism , Virulence Factors
19.
Am J Respir Cell Mol Biol ; 47(4): 445-53, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22582175

ABSTRACT

Antibiotics-induced release of the pore-forming virulence factor pneumolysin (PLY) in patients with pneumococcal pneumonia results in its presence days after lungs are sterile and is a major factor responsible for the induction of permeability edema. Here we sought to identify major mechanisms mediating PLY-induced endothelial dysfunction. We evaluated PLY-induced endothelial hyperpermeability in human lung microvascular endothelial cells (HL-MVECs) and human lung pulmonary artery endothelial cells in vitro and in mice instilled intratracheally with PLY. PLY increases permeability in endothelial monolayers by reducing stable and dynamic microtubule content and modulating VE-cadherin expression. These events, dependent upon an increased calcium influx, are preceded by protein kinase C (PKC)-α activation, perturbation of the RhoA/Rac1 balance, and an increase in myosin light chain phosphorylation. At later time points, PLY treatment increases the expression and activity of arginase in HL-MVECs. Arginase inhibition abrogates and suppresses PLY-induced endothelial barrier dysfunction by restoring NO generation. Consequently, a specific PKC-α inhibitor and the TNF-derived tonoplast intrinsic protein peptide, which blunts PLY-induced PKC-α activation, are able to prevent activation of arginase in HL-MVECs and to reduce PLY-induced endothelial hyperpermeability in mice. Arginase I (AI)(+/-)/arginase II (AII)(-/-) C57BL/6 mice, displaying a significantly reduced arginase I expression in the lungs, are significantly less sensitive to PLY-induced capillary leak than their wild-type or AI(+/+)/AII(-/-) counterparts, indicating an important role for arginase I in PLY-induced endothelial hyperpermeability. These results identify PKC-α and arginase I as potential upstream and downstream therapeutic targets in PLY-induced pulmonary endothelial dysfunction.


Subject(s)
Arginase/metabolism , Capillary Permeability , Endothelial Cells/metabolism , Lung/pathology , Protein Kinase C-alpha/metabolism , Streptolysins/pharmacology , Animals , Antigens, CD/metabolism , Arginase/antagonists & inhibitors , Bacterial Proteins/pharmacology , Cadherins/metabolism , Calcium Signaling , Cells, Cultured , Endothelial Cells/enzymology , Enzyme Inhibitors/pharmacology , Humans , Lung/blood supply , Lung/immunology , Male , Mice , Mice, Inbred C57BL , Microtubules/metabolism , Microvessels/pathology , Pneumonia/enzymology , Pneumonia/immunology , Pneumonia/pathology , Protein Kinase C-alpha/antagonists & inhibitors , rhoA GTP-Binding Protein/metabolism
20.
Proc Natl Acad Sci U S A ; 109(6): 2084-9, 2012 Feb 07.
Article in English | MEDLINE | ID: mdl-22308467

ABSTRACT

Aggressive treatment with antibiotics in patients infected with Streptococcus pneumoniae induces release of the bacterial virulence factor pneumolysin (PLY). Days after lungs are sterile, this pore-forming toxin can still induce pulmonary permeability edema in patients, characterized by alveolar/capillary barrier dysfunction and impaired alveolar liquid clearance (ALC). ALC is mainly regulated through Na(+) transport by the apically expressed epithelial sodium channel (ENaC) and the basolaterally expressed Na(+)/K(+)-ATPase in type II alveolar epithelial cells. Because no standard treatment is currently available to treat permeability edema, the search for novel therapeutic candidates is of high priority. We detected mRNA expression for the active receptor splice variant SV1 of the hypothalamic polypeptide growth hormone-releasing hormone (GHRH), as well as for GHRH itself, in human lung microvascular endothelial cells (HL-MVEC). Therefore, we have evaluated the effect of the GHRH agonist JI-34 on PLY-induced barrier and ALC dysfunction. JI-34 blunts PLY-mediated endothelial hyperpermeability in monolayers of HL-MVEC, in a cAMP-dependent manner, by means of reducing the phosphorylation of myosin light chain and vascular endothelial (VE)-cadherin. In human airway epithelial H441 cells, PLY significantly impairs Na(+) uptake, but JI-34 restores it to basal levels by means of increasing cAMP levels. Intratracheal instillation of PLY into C57BL6 mice causes pulmonary alveolar epithelial and endothelial hyperpermeability as well as edema formation, all of which are blunted by JI-34. These findings point toward a protective role of the GHRH signaling pathway in PLY-induced permeability edema.


Subject(s)
Growth Hormone-Releasing Hormone/agonists , Pulmonary Edema/pathology , Streptolysins/toxicity , Animals , Antigens, CD/metabolism , Bacterial Proteins/toxicity , Cadherins/metabolism , Endothelial Cells/metabolism , Endothelial Cells/pathology , Gene Expression Regulation , Growth Hormone-Releasing Hormone/genetics , Growth Hormone-Releasing Hormone/metabolism , Humans , Ion Channel Gating , Lung/metabolism , Lung/pathology , Mice , Mice, Inbred C57BL , Microvessels/pathology , Myosin Light Chains/metabolism , Permeability , Phosphorylation , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/pathology , Pulmonary Edema/genetics , Pulmonary Edema/physiopathology , RNA Splicing/genetics , Receptors, Neuropeptide/genetics , Receptors, Neuropeptide/metabolism , Receptors, Pituitary Hormone-Regulating Hormone/genetics , Receptors, Pituitary Hormone-Regulating Hormone/metabolism , Sodium Channels/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...