Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 604
Filter
Add more filters

Publication year range
1.
Mol Cell ; 81(1): 49-66.e8, 2021 01 07.
Article in English | MEDLINE | ID: mdl-33242393

ABSTRACT

Breathing depends on pulmonary surfactant, a mixture of phospholipids and proteins, secreted by alveolar type II cells. Surfactant requires lamellar bodies (LBs), organelles containing densely packed concentric membrane layers, for storage and secretion. LB biogenesis remains mysterious but requires surfactant protein B (SP-B), which is synthesized as a precursor (pre-proSP-B) that is cleaved during trafficking into three related proteins. Here, we elucidate the functions and cooperation of these proteins in LB formation. We show that the N-terminal domain of proSP-B is a phospholipid-binding and -transfer protein whose activities are required for proSP-B export from the endoplasmic reticulum (ER) and sorting to LBs, the conversion of proSP-B into lipoprotein particles, and neonatal viability in mice. The C-terminal domain facilitates ER export of proSP-B. The mature middle domain, generated after proteolytic cleavage of proSP-B, generates the striking membrane layers characteristic of LBs. Together, our results lead to a mechanistic model of LB biogenesis.


Subject(s)
Endoplasmic Reticulum/metabolism , Lipoproteins/metabolism , Multiprotein Complexes/metabolism , Pulmonary Surfactant-Associated Protein B/metabolism , Animals , Female , HEK293 Cells , Humans , Lipoproteins/chemistry , Mice , Multiprotein Complexes/chemistry , Protein Domains , Pulmonary Surfactant-Associated Protein B/chemistry
2.
Lung ; 202(3): 299-315, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38684519

ABSTRACT

PURPOSE: Acute respiratory distress syndrome (ARDS) is a major cause of hypoxemic respiratory failure in adults. In ARDS extensive inflammation and leakage of fluid into the alveoli lead to dysregulation of pulmonary surfactant metabolism and function. Altered surfactant synthesis, secretion, and breakdown contribute to the clinical features of decreased lung compliance and alveolar collapse. Lung function in ARDS could potentially be restored with surfactant replacement therapy, and synthetic surfactants with modified peptide analogues may better withstand inactivation in ARDS alveoli than natural surfactants. METHODS: This study aimed to investigate the activity in vitro and the bolus effect (200 mg phospholipids/kg) of synthetic surfactant CHF5633 with analogues of SP-B and SP-C, or natural surfactant Poractant alfa (Curosurf®, both preparations Chiesi Farmaceutici S.p.A.) in a severe ARDS model (the ratio of partial pressure arterial oxygen and fraction of inspired oxygen, P/F ratio ≤ 13.3 kPa) induced by hydrochloric acid instillation followed by injurious ventilation in adult New Zealand rabbits. The animals were ventilated for 4 h after surfactant treatment and the respiratory parameters, histological appearance of lung parenchyma and levels of inflammation, oxidative stress, surfactant dysfunction, and endothelial damage were evaluated. RESULTS: Both surfactant preparations yielded comparable improvements in lung function parameters, reductions in lung injury score, pro-inflammatory cytokines levels, and lung edema formation compared to untreated controls. CONCLUSIONS: This study indicates that surfactant replacement therapy with CHF5633 improves lung function and lung architecture, and attenuates inflammation in severe ARDS in adult rabbits similarly to Poractant alfa. Clinical trials have so far not yielded conclusive results, but exogenous surfactant may be a valid supportive treatment for patients with ARDS given its anti-inflammatory and lung-protective effects.


Subject(s)
Biological Products , Disease Models, Animal , Lung , Oxidative Stress , Phospholipids , Pulmonary Surfactant-Associated Protein B , Pulmonary Surfactant-Associated Protein C , Pulmonary Surfactants , Respiratory Distress Syndrome , Animals , Rabbits , Respiratory Distress Syndrome/drug therapy , Respiratory Distress Syndrome/physiopathology , Pulmonary Surfactants/pharmacology , Lung/drug effects , Lung/pathology , Lung/physiopathology , Lung/metabolism , Phospholipids/pharmacology , Biological Products/pharmacology , Biological Products/therapeutic use , Pulmonary Surfactant-Associated Protein B/pharmacology , Pulmonary Surfactant-Associated Protein B/metabolism , Oxidative Stress/drug effects , Pulmonary Surfactant-Associated Protein C/pharmacology , Male , Bronchoalveolar Lavage Fluid , Peptide Fragments , Phosphatidylcholines
3.
Int J Mol Sci ; 25(12)2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38928127

ABSTRACT

Valvular disease is a complex pathological condition that impacts countless individuals around the globe. Due to limited treatments, it is crucial to understand its mechanisms to identify new targets. Valve disease may result in pulmonary venous hypertension, which is linked to compromised functioning of the alveolar and capillary membranes and hindered gas exchange. Nonetheless, the correlation between surfactant proteins (SPs) and valve disease remains unexplored. A total of 44 patients were enrolled in this study, with 36 undergoing aortic valve replacement and 8 needing a second aortic valve substitution due to bioprosthetic valve degeneration. Ten healthy subjects were also included. The results showed that patients who underwent both the first valve replacement and the second surgery had significantly higher levels of immature SP-B (proSP-B) compared to control subjects. The levels of the extra-lung collectin SP-D were higher in patients who needed a second surgery due to bioprosthetic valve degeneration, while SP-A levels remained unchanged. The research also showed that there was no reciprocal relationship between inflammation and SP-D as the levels of inflammatory mediators did not differ between groups. The present study demonstrates that circulating proSP-B serves as a reliable marker of alveolar-capillary membrane damage in patients with valvular heart disease.


Subject(s)
Aortic Valve Stenosis , Aortic Valve , Calcinosis , Pulmonary Surfactant-Associated Protein B , Humans , Aortic Valve Stenosis/blood , Aortic Valve Stenosis/surgery , Male , Female , Pulmonary Surfactant-Associated Protein B/blood , Pulmonary Surfactant-Associated Protein B/metabolism , Aged , Calcinosis/blood , Aortic Valve/surgery , Aortic Valve/pathology , Middle Aged , Biomarkers/blood , Case-Control Studies
4.
Langmuir ; 39(12): 4338-4350, 2023 03 28.
Article in English | MEDLINE | ID: mdl-36917773

ABSTRACT

The lining of the alveoli is covered by pulmonary surfactant, a complex mixture of surface-active lipids and proteins that enables efficient gas exchange between inhaled air and the circulation. Despite decades of advancements in the study of the pulmonary surfactant, the molecular scale behavior of the surfactant and the inherent role of the number of different lipids and proteins in surfactant behavior are not fully understood. The most important proteins in this complex system are the surfactant proteins SP-B and SP-C. Given this, in this work we performed nonequilibrium all-atom molecular dynamics simulations to study the interplay of SP-B and SP-C with multicomponent lipid monolayers mimicking the pulmonary surfactant in composition. The simulations were complemented by z-scan fluorescence correlation spectroscopy and atomic force microscopy measurements. Our state-of-the-art simulation model reproduces experimental pressure-area isotherms and lateral diffusion coefficients. In agreement with previous research, the inclusion of either SP-B and SP-C increases surface pressure, and our simulations provide a molecular scale explanation for this effect: The proteins display preferential lipid interactions with phosphatidylglycerol, they reside predominantly in the lipid acyl chain region, and they partition into the liquid expanded phase or even induce it in an otherwise packed monolayer. The latter effect is also visible in our atomic force microscopy images. The research done contributes to a better understanding of the roles of specific lipids and proteins in surfactant function, thus helping to develop better synthetic products for surfactant replacement therapy used in the treatment of many fatal lung-related injuries and diseases.


Subject(s)
Pulmonary Surfactants , Biophysical Phenomena , Phospholipids/chemistry , Proteins , Pulmonary Surfactant-Associated Protein B/chemistry , Pulmonary Surfactants/chemistry , Surface Properties , Surface-Active Agents , Pulmonary Surfactant-Associated Protein C/chemistry
5.
J Lipid Res ; 63(4): 100196, 2022 04.
Article in English | MEDLINE | ID: mdl-35300983

ABSTRACT

Atherosclerotic CVD is the major cause of death in patients with type 1 diabetes mellitus (T1DM). Alterations in the HDL proteome have been shown to associate with prevalent CVD in T1DM. We therefore sought to determine which proteins carried by HDL might predict incident CVD in patients with T1DM. Using targeted MS/MS, we quantified 50 proteins in HDL from 181 T1DM subjects enrolled in the prospective Coronary Artery Calcification in Type 1 Diabetes study. We used Cox proportional regression analysis and a case-cohort design to test associations of HDL proteins with incident CVD (myocardial infarction, coronary artery bypass grafting, angioplasty, or death from coronary heart disease). We found that only one HDL protein-SFTPB (pulmonary surfactant protein B)-predicted incident CVD in all the models tested. In a fully adjusted model that controlled for lipids and other risk factors, the hazard ratio was 2.17 per SD increase of SFTPB (95% confidence interval, 1.12-4.21, P = 0.022). In addition, plasma fractionation demonstrated that SFTPB is nearly entirely bound to HDL. Although previous studies have shown that high plasma levels of SFTPB associate with prevalent atherosclerosis only in smokers, we found that SFTPB predicted incident CVD in T1DM independently of smoking status and a wide range of confounding factors, including HDL-C, LDL-C, and triglyceride levels. Because SFTPB is almost entirely bound to plasma HDL, our observations support the proposal that SFTPB carried by HDL is a marker-and perhaps mediator-of CVD risk in patients with T1DM.


Subject(s)
Atherosclerosis , Cardiovascular Diseases , Diabetes Mellitus, Type 1 , Pulmonary Surfactant-Associated Protein B , Cholesterol, HDL , Diabetes Mellitus, Type 1/complications , Humans , Prospective Studies , Risk Factors , Tandem Mass Spectrometry
6.
Environ Toxicol ; 37(9): 2291-2301, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35689653

ABSTRACT

Exposure to silica nanoparticles (SiNPs) is related to the dysregulation of pulmonary surfactant that maintains lung stability and function. Nevertheless, there are limited studies concerning the interaction and influence between SiNPs and pulmonary surfactant, and the damage and mechanism are still unclear. Herein, we used A549 cells to develop an in vitro model, with which we investigated the effect of SiNPs exposure on the expression of pulmonary surfactant and the potential regulatory mechanism. The results showed that SiNPs were of cytotoxicity in regarding of reduced cell viability and promoted the production of excessive reactive oxygen species (ROS). Additionally, the JNK/c-Jun signaling pathway was activated, and the expression of surfactant protein A (SP-A) and surfactant protein B (SP-B) was decreased. After the cells being treated with N-acetyl-L-cysteine (NAC), we found that the ROS content was effectively downregulated, and the expression of proteins related to JNK and c-Jun signaling pathways was suppressed. In contrast, the expression of SP-A and SP-B was enhanced. Furthermore, we treated the cells with JNK inhibitor and c-Jun-siRNA and found that the expression of protein related to JNK and c-Jun signaling pathways, as well as SP-A and SP-B, changed in line with that of NAC treatment. These findings suggest that SiNPs exposure can upregulate ROS and activate the JNK/c-Jun signaling pathway in A549 cells, thereby inhibiting the expression of SP-A and SP-B proteins.


Subject(s)
Lung , Nanoparticles , Pulmonary Surfactant-Associated Protein A , Pulmonary Surfactant-Associated Protein B , Silicon Dioxide , A549 Cells , Acetylcysteine/pharmacology , Apoptosis , Genes, jun/genetics , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Lung/metabolism , Nanoparticles/toxicity , Pulmonary Surfactant-Associated Protein A/metabolism , Pulmonary Surfactant-Associated Protein B/metabolism , Pulmonary Surfactants/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Silicon Dioxide/toxicity
7.
Biophys J ; 120(2): 243-253, 2021 01 19.
Article in English | MEDLINE | ID: mdl-33347885

ABSTRACT

To determine how different constituents of pulmonary surfactant affect its phase behavior, we measured wide-angle x-ray scattering (WAXS) from oriented bilayers. Samples contained the nonpolar and phospholipids (N&PL) obtained from calf lung surfactant extract (CLSE), which also contains the hydrophobic surfactant proteins SP-B and SP-C. Mixtures with different ratios of N&PL and CLSE provided the same set of lipids with different amounts of the proteins. At 37°C, N&PL by itself forms coexisting Lα and Lß phases. In the Lß structure, the acyl chains of the phospholipids occupy an ordered array that has melted by 40°C. This behavior suggests that the Lß composition is dominated by dipalmitoyl phosphatidylcholine (DPPC), which is the most prevalent component of CLSE. The Lß chains, however, lack the tilt of the Lß' phase formed by pure DPPC. At 40°C, WAXS also detects an additional diffracted intensity, the location of which suggests a correlation among the phospholipid headgroups. The mixed samples of N&PL with CLSE show that increasing amounts of the proteins disrupt both the Lß phase and the headgroup correlation. With physiological levels of the proteins in CLSE, both types of order are absent. These results with bilayers at physiological temperatures indicate that the hydrophobic surfactant proteins disrupt the ordered structures that have long been considered essential for the ability of pulmonary surfactant to sustain low surface tensions. They agree with prior fluorescence micrographic results from monomolecular films of CLSE, suggesting that at physiological temperatures, any ordered phase is likely to be absent or occupy a minimal interfacial area.


Subject(s)
Pulmonary Surfactants , 1,2-Dipalmitoylphosphatidylcholine , Phospholipids , Proteins , Pulmonary Surfactant-Associated Protein B , Surface Tension
8.
Exp Physiol ; 106(5): 1303-1311, 2021 05.
Article in English | MEDLINE | ID: mdl-33729612

ABSTRACT

NEW FINDINGS: What is the central question of this study? It is reported that polymorphism of the gene for pulmonary surfactant-associated protein B (SFTPB) is associated with chronic obstructive pulmonary disease (COPD): what are the function and mechanism of action of SFTPB in COPD? What is the main finding and its importance? Under stimulation of the risk factors of COPD, SFTPB expression is decreased, which may be involved in the formation of COPD. The progress of COPD induces an inflammatory response and reduces SFTPB expression. Levels of prostaglandin-endoperoxide synthase-2 (PTGS2) and inflammatory responses are changed by SFTPB, which indicates that SFTPB promotes the progression of COPD by PTGS2 and inflammation. ABSTRACT: Pulmonary surfactant-associated protein B (SFTPB) is a critical protein for lung homeostasis, and polymorphism of its gene is associated with chronic obstructive pulmonary disease (COPD). However, few studies have so far confirmed the functional involvement of SFTPB in COPD. Serum SFTPB and inflammatory cytokine levels were measured in 54 patients with acute exacerbation of COPD and 29 healthy controls. A549 cells were induced using 10% cigarette smoke extract (CSE) and treated with dexamethasone to investigate the effect of inflammation on SFTPB expression, and the effect of SFTPB overexpression and silencing on inflammatory cytokines was measured using real-time PCR and enzyme-linked immunosorbent assay. SFTPB expression was assessed in mouse lung tissues using immunofluorescence. Serum levels of SFTPB were significantly lower in COPD patients than in controls (P = 0.009). Conversely, levels of interleukin (IL)-6 and prostaglandin-endoperoxide synthase-2 (PTGS2) were increased in COPD patients (IL-6: P = 0.006; PTGS2: P = 0.043). After CSE treatment, SFTPB mRNA and protein levels were significantly decreased compared to controls (mRNA: P = 0.002; protein: P = 0.011), while IL-6, IL-8 and PTGS2 were elevated. Dexamethasone treatment increased SFTPB levels. Following overexpression of SFTPB in A549 cells, mRNA and protein levels of IL-6, IL-8 and PTGS2 were significantly reduced, while gene silencing induced the opposite effect. SFTPB levels were significantly reduced in the lung tissue of a mouse model of COPD compared to controls. Reduced SFTPB levels may induce PTGS2 and inflammatory responses in COPD and SFTPB could be a key protein for evaluation of COPD progression.


Subject(s)
Cyclooxygenase 2/blood , Pulmonary Disease, Chronic Obstructive , Pulmonary Surfactant-Associated Protein B , A549 Cells , Animals , Humans , Inflammation , Lung/metabolism , Mice , Protein Precursors , Pulmonary Surfactant-Associated Protein B/blood , Pulmonary Surfactant-Associated Protein B/genetics , Pulmonary Surfactant-Associated Proteins
9.
BMC Pulm Med ; 21(1): 330, 2021 Oct 22.
Article in English | MEDLINE | ID: mdl-34686153

ABSTRACT

BACKGROUND: Optimal functionality of synthetic lung surfactant for treatment of respiratory distress syndrome in preterm infants largely depends on the quality and quantity of the surfactant protein B (SP-B) peptide mimic and the lipid mixture. B-YL peptide is a 41-residue sulfur-free SP-B mimic with its cysteine and methionine residues replaced by tyrosine and leucine, respectively, to enhance its oxidation resistance. AIM: Testing the structural and functional stability of the B-YL peptide in synthetic surfactant lipids after long-term storage. METHODS: The structural and functional properties of B-YL peptide in surfactant lipids were studied using three production runs of B-YL peptides in synthetic surfactant lipids. Each run was held at 5 °C ambient temperature for three years and analyzed with structural and computational techniques, i.e., MALDI-TOF mass spectrometry, ATR-Fourier Transform Infrared Spectroscopy (ATR-FTIR), secondary homology modeling of a preliminary B-YL structure, and tertiary Molecular Dynamic simulations of B-YL in surfactant lipids, and with functional methods, i.e., captive bubble surfactometry (CBS) and retesting in vivo surface activity in surfactant-deficient young adult rabbits. RESULTS: MALDI-TOF mass spectrometry showed no degradation of the B-YL peptide as a function of stored time. ATR-FTIR studies demonstrated that the B-YL peptide still assumed stable alpha-helical conformations in synthetic surfactant lipids. These structural findings correlated with excellent in vitro surface activity during both quasi-static and dynamic cycling on CBS after three years of cold storage and in vivo surface activity of the aged formulations with improvements in oxygenation and dynamic lung compliance approaching those of the positive control surfactant Curosurf®. CONCLUSIONS: The structure of the B-YL peptide and the in vitro and in vivo functions of the B-YL surfactant were each maintained after three years of refrigeration storage.


Subject(s)
Pulmonary Surfactant-Associated Protein B/chemistry , Pulmonary Surfactants/chemistry , Surface-Active Agents/chemistry , Animals , Drug Stability , Lipid Metabolism , Pulmonary Surfactant-Associated Protein B/metabolism , Pulmonary Surfactants/metabolism , Rabbits , Surface-Active Agents/metabolism
10.
J Pediatr ; 225: 90-96.e1, 2020 10.
Article in English | MEDLINE | ID: mdl-32553868

ABSTRACT

OBJECTIVE: To compare efficacy and safety of a new synthetic surfactant, CHF5633, enriched with surfactant proteins, SP-B and SP-C peptide analogues, with porcine surfactant, poractant alfa, for the treatment of respiratory distress syndrome in infants born preterm. STUDY DESIGN: Neonates born preterm on respiratory support requiring fraction of inspired oxygen (FiO2) ≥0.30 from 240/7 to 266/7 weeks and FiO2 ≥0.35 from 270/7 to 296/7 weeks of gestation to maintain 88%-95% oxygen saturation were randomized to receive 200 mg/kg of CHF5633 or poractant alfa. If necessary, redosing was given at 100 mg/kg. Efficacy end points were oxygen requirement (FiO2, respiratory severity score [FiO2 × mean airway pressure]) in the first 24 hours, 7 and 28 days, discharge home, and/or 36 weeks of postmenstrual age; mortality and bronchopulmonary dysplasia at 28 days and 36 weeks of PMA. Adverse events and immunogenicity were monitored for safety. RESULTS: Of the 123 randomized neonates, 113 were treated (56 and 57 in CHF5633 and poractant alfa groups, respectively). In both arms, FiO2 and respiratory severity score decreased from baseline at all time points (P < .001) with no statistically significant differences between groups. Rescue surfactant use (19 [33.9%] vs 17 [29.8%]), bronchopulmonary dysplasia (31 [55.4%] and 32 [56.1%]), and mortality at day 28 (4 [7.1%] and 3 [5.3%]) were similar in the CHF5633 and poractant alfa groups, respectively. In 2 (3.4%) and 1 (1.7%) neonates, adverse drug reactions were reported in CHF5633 and poractant alfa groups, respectively. No immunogenicity was detected. CONCLUSIONS: Treatment with CHF5633 showed similar efficacy and safety as poractant alfa in neonates born preterm with moderate-to-severe respiratory distress syndrome. TRIAL REGISTRATION: ClinicalTrials.gov: NCT02452476.


Subject(s)
Biological Products/therapeutic use , Peptide Fragments/therapeutic use , Phosphatidylcholines/therapeutic use , Phospholipids/therapeutic use , Pulmonary Surfactant-Associated Protein B/therapeutic use , Pulmonary Surfactant-Associated Protein C/therapeutic use , Pulmonary Surfactants/therapeutic use , Respiratory Distress Syndrome, Newborn/drug therapy , Biomarkers/metabolism , Bronchopulmonary Dysplasia/drug therapy , Double-Blind Method , Female , Humans , Infant, Newborn , Infant, Premature , Male , Oxygen/therapeutic use , Treatment Outcome
11.
Am J Obstet Gynecol ; 223(6): 921.e1-921.e10, 2020 12.
Article in English | MEDLINE | ID: mdl-32445634

ABSTRACT

BACKGROUND: Administration of antenatal steroids is standard of care for women assessed to be at imminent risk of preterm delivery. There is a marked variation in antenatal steroid dosing strategy, selection for treatment criteria, and agent choice worldwide. This, combined with very limited optimization of antenatal steroid use per se, means that treatment efficacy is highly variable, and the rate of respiratory distress syndrome is decreased to perhaps as low as 40%. In some cases, antenatal steroid use is associated with limited benefit and potential harm. OBJECTIVE: We hypothesized that individual differences in maternofetal steroid exposure would contribute to observed variability in antenatal steroid treatment efficacy. Using a chronically catheterized sheep model of pregnancy, we aimed to explore the relationship between maternofetal steroid exposure and antenatal steroid treatment efficacy as determined by functional lung maturation in preterm lambs undergoing ventilation. STUDY DESIGN: Ewes carrying a single fetus underwent surgery to catheterize a fetal and maternal jugular vein at 119 days' gestation. Animals recovered for 24 hours before being randomized to either (1) a single maternal intramuscular injection of 2 mL saline (negative control group, n=10) or (2) a single maternal intramuscular injection of 0.25 mg/kg betamethasone phosphate plus acetate (antenatal steroid group, n=20). Serial maternal and fetal plasma samples were collected from each animal after 48 hours before fetuses were delivered and ventilated for 30 minutes. Total and free plasma betamethasone concentration was measured by mass spectrometry. Fetal lung tissue was collected for analysis using quantitative polymerase chain reaction. RESULTS: One animal from the control group and one animal from the antenatal steroid group did not complete their treatment protocol and were removed from analyses. Animals in the antenatal steroid group were divided into a responder subgroup (n=12/19) and a nonresponder subgroup (n=7/19) using a cutoff of partial pressure of arterial CO2 at 30-minute ventilation within 2 standard deviations of the mean value from saline-treated negative control group animals. Although antenatal steroid improved fetal lung maturation in the undivided antenatal steroid group and in the responder subgroup both physiologically (blood gas- and ventilation-related data) and biochemically (messenger ribonucleic acid expression related to fetal lung maturation), these values did not improve relative to saline-treated control group animals in the antenatal steroid nonresponder subgroup. No differences in betamethasone distribution, clearance, or protein binding were identified between the antenatal steroid responder and nonresponder subgroups. CONCLUSION: This study correlated individual maternofetal steroid exposures with preterm lung maturation as determined by pulmonary ventilation. Herein, approximately 40% of preterm lambs exposed to antenatal steroids had lung maturation that was not significantly different to saline-treated control group animals. These nonresponsive animals received maternal and fetal betamethasone exposures identical to animals that had a significant improvement in functional lung maturation. These data suggest that the efficacy of antenatal steroid therapy is not solely determined by maternofetal drug levels and that individual fetal or maternal factors may play a role in determining treatment outcomes in response to glucocorticoid signaling.


Subject(s)
Betamethasone/analogs & derivatives , Fetal Organ Maturity/drug effects , Glucocorticoids/pharmacology , Lung/drug effects , Animals , Aquaporin 1/drug effects , Aquaporin 1/genetics , Aquaporin 5/drug effects , Aquaporin 5/genetics , Betamethasone/blood , Betamethasone/pharmacology , Blood Gas Analysis , Carbon Dioxide , Epithelial Sodium Channels/drug effects , Epithelial Sodium Channels/genetics , Female , Fetal Organ Maturity/genetics , Glucocorticoids/blood , Lung/metabolism , Lung/physiopathology , Lung Compliance/drug effects , Mass Spectrometry , Maternal-Fetal Exchange , Partial Pressure , Perinatal Care , Polymerase Chain Reaction , Pregnancy , Premature Birth , Prenatal Care , Pulmonary Surfactant-Associated Protein A/drug effects , Pulmonary Surfactant-Associated Protein A/genetics , Pulmonary Surfactant-Associated Protein B/drug effects , Pulmonary Surfactant-Associated Protein B/genetics , Pulmonary Surfactant-Associated Protein C/drug effects , Pulmonary Surfactant-Associated Protein C/genetics , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Random Allocation , Respiration, Artificial , Sheep
12.
Pediatr Res ; 87(3): 511-517, 2020 02.
Article in English | MEDLINE | ID: mdl-30776794

ABSTRACT

BACKGROUND: Current in vitro human lung epithelial cell models derived from adult tissues may not accurately represent all attributes that define homeostatic and disease mechanisms relevant to the pediatric lung. METHODS: We report methods for growing and differentiating primary Pediatric Human Lung Epithelial (PHLE) cells from organ donor infant lung tissues. We use immunohistochemistry, flow cytometry, quantitative RT-PCR, and single cell RNA sequencing (scRNAseq) analysis to characterize the cellular and transcriptional heterogeneity of PHLE cells. RESULTS: PHLE cells can be expanded in culture up to passage 6, with a doubling time of ~4 days, and retain attributes of highly enriched epithelial cells. PHLE cells can form resistant monolayers, and undergo differentiation when placed at air-liquid interface. When grown at Air-Liquid Interface (ALI), PHLE cells expressed markers of airway epithelial cell lineages. scRNAseq suggests the cultures contained 4 main sub-phenotypes defined by expression of FOXJ1, KRT5, MUC5B, and SFTPB. These cells are available to the research community through the Developing Lung Molecular Atlas Program Human Tissue Core. CONCLUSION: Our data demonstrate that PHLE cells provide a novel in vitro human cell model that represents the pediatric airway epithelium, which can be used to study perinatal developmental and pediatric disease mechanisms.


Subject(s)
Cell Separation , Epithelial Cells/physiology , Lung/cytology , Tissue Donors , Age Factors , Cell Differentiation , Cell Lineage , Cell Proliferation , Cells, Cultured , Epithelial Cells/metabolism , Epithelial Cells/virology , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Host-Pathogen Interactions , Humans , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza, Human/genetics , Influenza, Human/metabolism , Influenza, Human/virology , Keratin-5/genetics , Keratin-5/metabolism , Mucin-5B/genetics , Mucin-5B/metabolism , Phenotype , Primary Cell Culture , Pulmonary Surfactant-Associated Protein B/genetics , Pulmonary Surfactant-Associated Protein B/metabolism , RNA-Seq , Single-Cell Analysis
13.
Phys Chem Chem Phys ; 22(27): 15231-15241, 2020 Jul 21.
Article in English | MEDLINE | ID: mdl-32608403

ABSTRACT

Lung surfactant (LS) monolayers that continuously expand and compress during breathing cycles, act as the first line barrier for inhaled nanoparticles. It is known that nanoparticles which adsorb to the surface of the surfactant layer facilitate the rearrangement of lipids and peptides at various stages of the breathing cycle. However, the structural mechanisms for this ability of the lipid rearrangement are not yet fully understood. Coarse-grained molecular dynamics simulations are performed to investigate the role of surfactant protein B (SP-B) segments (SP-B1-25) in modulating the biophysical properties of the surfactant monolayer in the presence of polydisperse gold nanoparticles (AuNPs) at different concentrations. Herein, we observe that the AuNPs significantly alter the inherent structural and dynamical properties of the monolayer and its components in three different breathing states. When adsorbed into the monolayer, the AuNPs inhibit the ability of the monolayer to recover its surface tension and other properties. The presence of SP-B1-25 in the monolayer accelerates the diffusion of the monolayer phospholipids, contrarily to the role of AuNPs on phospholipid diffusion. Also, the AuNPs and the peptides in the monolayer significantly increase their agglomeration in the presence of one another. Overall, the simulations predict that the presence of polydisperse AuNPs hampers the stability and biophysical functions of the LS in contrast to the role of the peptide. This study provides a clear view of the hydrophobic peptide role in the LS monolayer at the interface along with the interactions and the translocation of AuNPs that could have a significant impact to assess the NPs inhalation.


Subject(s)
Gold/chemistry , Metal Nanoparticles/chemistry , Molecular Dynamics Simulation , Pulmonary Surfactant-Associated Protein B/chemistry , Protein Conformation
14.
Am J Respir Crit Care Med ; 200(7): 881-887, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31002528

ABSTRACT

Rationale: Pulmonary alveolar proteinosis (PAP) is characterized by filling of the alveolar spaces by lipoprotein-rich material of ill-defined composition, and is caused by molecularly different and often rare diseases that occur from birth to old age.Objectives: To perform a quantitative lipidomic analysis of lipids and the surfactant proteins A, B, and C in lavage fluids from patients with proteinosis of different causes in comparison with healthy control subjects.Methods: During the last two decades, we have collected BAL samples from patients with PAP due to autoantibodies against granulocyte-macrophage colony-stimulating factor; genetic mutations in CSF2RA (colony-stimulating factor 2 receptor α-subunit), MARS (methionyl aminoacyl-tRNA synthetase), FARSB (phenylalanine-tRNA synthetase, ß-subunit), and NPC2 (Niemann-Pick disease type C2); and secondary to myeloid leukemia. Their lipid composition was quantified.Measurements and Main Results: Free cholesterol was largely increased by 60-fold and cholesteryl esters were increased by 24-fold. There was an excessive, more than 130-fold increase in ceramide and other sphingolipids. In particular, the long-chain ceramides d18:1/20:0 and d18:1/24:0 were elevated and likely contributed to the proapoptotic environment observed in PAP. Cellular debris lipids such as phosphatidylethanolamine and phosphatidylserine were only moderately increased, by four- to sevenfold. The surfactant lipid class phosphatidylcholine expanded 17-fold, lysophosphatidylcholine expanded 54-fold, and the surfactant proteins A, B, and C expanded 144-, 4-, and 17-fold, respectively. These changes did not differ among the various diseases that cause PAP.Conclusions: This insight into the alveolar lipidome may provide monitoring tools and lead to new therapeutic strategies for PAP.


Subject(s)
Lipid Metabolism , Lipidomics , Pulmonary Alveolar Proteinosis/metabolism , Pulmonary Surfactant-Associated Proteins/metabolism , Adolescent , Adult , Apoptosis , Autoimmune Diseases/metabolism , Bronchoalveolar Lavage Fluid , Case-Control Studies , Ceramides/metabolism , Child , Child, Preschool , Cholesterol/metabolism , Cholesterol Esters/metabolism , Female , Genetic Diseases, X-Linked/genetics , Genetic Diseases, X-Linked/metabolism , Humans , Infant , Leukemia, Myeloid/complications , Male , Methionine-tRNA Ligase/genetics , Middle Aged , Phenylalanine-tRNA Ligase/genetics , Phosphatidylcholines/metabolism , Phosphatidylethanolamines/metabolism , Phosphatidylserines/metabolism , Pulmonary Alveolar Proteinosis/etiology , Pulmonary Alveolar Proteinosis/genetics , Pulmonary Surfactant-Associated Protein A/metabolism , Pulmonary Surfactant-Associated Protein B/metabolism , Pulmonary Surfactant-Associated Protein C/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Sphingolipids/metabolism , Vesicular Transport Proteins/genetics , Young Adult
15.
J Biol Chem ; 293(24): 9399-9411, 2018 06 15.
Article in English | MEDLINE | ID: mdl-29700110

ABSTRACT

Pulmonary surfactant is a lipid/protein mixture that reduces surface tension at the respiratory air-water interface in lungs. Among its nonlipidic components are pulmonary surfactant-associated proteins B and C (SP-B and SP-C, respectively). These highly hydrophobic proteins are required for normal pulmonary surfactant function, and whereas past literature works have suggested possible SP-B/SP-C interactions and a reciprocal modulation effect, no direct evidence has been yet identified. In this work, we report an extensive fluorescence spectroscopy study of both intramolecular and intermolecular SP-B and SP-C interactions, using a combination of quenching and FRET steady-state and time-resolved methodologies. These proteins are compartmentalized in full surfactant membranes but not in pure 1-palmitoyl-2-oleoylphosphatidylcholine (POPC) vesicles, in accordance with their previously described preference for liquid disordered phases. From the observed static self-quenching and homo-FRET of BODIPY-FL labeled SP-B, we conclude that this protein forms homoaggregates at low concentration (lipid:protein ratio, 1:1000). Increases in polarization of BODIPY-FL SP-B and steady-state intensity of WT SP-B were observed upon incorporation of under-stoichiometric amounts of WT SP-C. Conversely, Marina Blue-labeled SP-C is quenched by over-stoichiometric amounts of WT SP-B, whereas under-stoichiometric concentrations of the latter actually increase SP-C emission. Time-resolved hetero-FRET from Marina Blue SP-C to BODIPY-FL SP-B confirm distinct protein aggregation behaviors with varying SP-B concentration. Based on these multiple observations, we propose a model for SP-B/SP-C interactions, where SP-C might induce conformational changes on SP-B complexes, affecting its aggregation state. The conclusions inferred from the present work shed light on the synergic functionality of both proteins in the pulmonary surfactant system.


Subject(s)
Lipid Bilayers/metabolism , Phospholipids/metabolism , Protein Interaction Maps , Pulmonary Surfactant-Associated Protein B/metabolism , Pulmonary Surfactant-Associated Protein C/metabolism , Animals , Fluorescence Polarization , Fluorescence Resonance Energy Transfer , Hydrophobic and Hydrophilic Interactions , Protein Aggregates , Protein Multimerization , Pulmonary Surfactant-Associated Protein B/chemistry , Pulmonary Surfactant-Associated Protein C/chemistry , Swine
16.
J Cell Biochem ; 120(9): 14804-14812, 2019 09.
Article in English | MEDLINE | ID: mdl-31016788

ABSTRACT

Plasma pro-surfactant protein B (pro-SFTPB) and N1,N12-diacetylspermine (DAS) can be used as markers for the diagnosis of non-small-cell lung carcinoma (NSCLC). Whether the genetic diversity affects the application value of Pro-SFTPB and DAS as a diagnostic marker for NSCLC is still unknown. This study aims to explore the relationship between SFTPB rs7316, rs9752 and PAOX rs1046175 gene polymorphisms and the diagnostic value of plasma Pro-SFTPB and DAS in patients with Chinese Han lung cancer. SFTPB rs7316, rs9752 and PAOX rs1046175 genotypes were analyzed by direct sequencing in 425 patients with NSCLC and 425 controls, and the levels of Pro-SFTPB and DAS in plasma were determined by enzyme-linked immunosorbent assay (ELISA). The area under the curve (AUC) of the SFTPB rs7316 locus TT genotype for the diagnosis of NSCLC was 0.758, and the AUC of the TC/CC genotype for the diagnosis of NSCLC was 0.872. The AUC of the SFTPB rs9752 locus GG genotype for the diagnosis of NSCLC was 0.935, and the AUC of the GC/CC genotype for the diagnosis of NSCLC was 0.648. The AUC of the PAOX rs1046175 locus GG for the diagnosis of NSCLC was 0.669, and the AUC of the GC/CC genotype for the diagnosis of NSCLC was 0.749. In conclusion, SFTPB rs7316, rs9752, and PAOX rs1046175 gene polymorphisms affect the diagnostic value of plasma Pro-SFTPB and DAS in patients with Chinese Han NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/diagnosis , Lung Neoplasms/diagnosis , Oxidoreductases Acting on CH-NH Group Donors/genetics , Polymorphism, Single Nucleotide , Protein Precursors/blood , Pulmonary Surfactant-Associated Protein B/genetics , Pulmonary Surfactant-Associated Proteins/blood , Spermine/analogs & derivatives , Adenocarcinoma of Lung/blood , Adenocarcinoma of Lung/diagnosis , Adenocarcinoma of Lung/genetics , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/analysis , Carcinoma, Non-Small-Cell Lung/blood , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Squamous Cell/blood , Carcinoma, Squamous Cell/diagnosis , Carcinoma, Squamous Cell/genetics , Case-Control Studies , Female , Follow-Up Studies , Humans , Lung Neoplasms/blood , Lung Neoplasms/genetics , Lymphatic Metastasis , Male , Middle Aged , Prognosis , Risk Factors , Spermine/blood
17.
J Intern Med ; 285(2): 165-186, 2019 02.
Article in English | MEDLINE | ID: mdl-30357986

ABSTRACT

Treatment of neonatal respiratory distress syndrome (RDS) using animal-derived lung surfactant preparations has reduced the mortality of handling premature infants with RDS to a 50th of that in the 1960s. The supply of animal-derived lung surfactants is limited and only a part of the preterm babies is treated. Thus, there is a need to develop well-defined synthetic replicas based on key components of natural surfactant. A synthetic product that equals natural-derived surfactants would enable cost-efficient production and could also facilitate the development of the treatments of other lung diseases than neonatal RDS. Recently the first synthetic surfactant that contains analogues of the two hydrophobic surfactant proteins B (SP-B) and SP-C entered clinical trials for the treatment of neonatal RDS. The development of functional synthetic analogues of SP-B and SP-C, however, is considerably more challenging than anticipated 30 years ago when the first structural information of the native proteins became available. For SP-B, a complex three-dimensional dimeric structure stabilized by several disulphides has necessitated the design of miniaturized analogues. The main challenge for SP-C has been the pronounced amyloid aggregation propensity of its transmembrane region. The development of a functional non-aggregating SP-C analogue that can be produced synthetically was achieved by designing the amyloidogenic native sequence so that it spontaneously forms a stable transmembrane α-helix.


Subject(s)
Infant, Premature , Lung Diseases/drug therapy , Pulmonary Surfactant-Associated Protein B/chemistry , Pulmonary Surfactant-Associated Protein C/chemistry , Pulmonary Surfactants/therapeutic use , Respiratory Distress Syndrome, Newborn/drug therapy , Humans , Infant, Newborn , Pulmonary Surfactants/chemistry
18.
Respir Res ; 20(1): 175, 2019 Aug 05.
Article in English | MEDLINE | ID: mdl-31382955

ABSTRACT

BACKGROUND: The amount of surfactant deposited in the lungs and its overall pulmonary distribution determine the therapeutic outcome of surfactant replacement therapy. Most of the currently available methods to determine the intrapulmonary distribution of surfactant are time-consuming and require surfactant labelling. Our aim was to assess the potential of Mass Spectrometry Imaging (MSI) as a label-free technique to qualitatively and quantitatively evaluate the distribution of surfactant to the premature lamb. METHODS: Twelve preterm lambs (gestational age 126-127d, term ~150d) were allocated in two experimental groups. Seven lambs were treated with an intratracheal bolus of the synthetic surfactant CHF5633 (200 mg/kg) and 5 lambs were managed with mechanical ventilation for 120 min, as controls. The right lung lobes of all lambs were gradually frozen while inflated to 20 cmH2O pressure for lung cryo-sections for MSI analysis. The intensity signals of SP-C analog and SP-B analog, the two synthetic peptides contained in the CHF5633 surfactant, were used to locate, map and quantify the intrapulmonary exogenous surfactant. RESULTS: Surfactant treatment was associated with a significant improvement of the mean arterial oxygenation and lung compliance (p < 0.05). Nevertheless, the physiological response to surfactant treatment was not uniform across all animals. SP-C analog and SP-B analog were successfully imaged and quantified by means of MSI in the peripheral lungs of all surfactant-treated animals. The intensity of the signal was remarkably low in untreated lambs, corresponding to background noise. The signal intensity of SP-B analog in each surfactant-treated animal, which represents the surfactant distributed to the peripheral right lung, correlated well with the physiologic response as assessed by the area under the curves of the individual arterial partial oxygen pressure and dynamic lung compliance curves of the lambs. CONCLUSIONS: Applying MSI, we were able to detect, locate and quantify the amount of exogenous surfactant distributed to the lower right lung of surfactant-treated lambs. The distribution pattern of SP-B analog correlated well with the pulmonary physiological outcomes of the animals. MSI is a valuable label-free technique which is able to simultaneously evaluate qualitative and quantitative drug distribution in the lung.


Subject(s)
Lung/metabolism , Peptide Fragments/analysis , Peptide Fragments/metabolism , Phosphatidylcholines/analysis , Phosphatidylcholines/metabolism , Pulmonary Surfactant-Associated Protein B/analysis , Pulmonary Surfactant-Associated Protein B/metabolism , Pulmonary Surfactant-Associated Protein C/analysis , Pulmonary Surfactant-Associated Protein C/metabolism , Pulmonary Surfactants/analysis , Pulmonary Surfactants/metabolism , Animals , Animals, Newborn , Lung/drug effects , Mass Spectrometry/methods , Peptide Fragments/pharmacology , Phosphatidylcholines/pharmacology , Pulmonary Surfactant-Associated Protein B/pharmacology , Pulmonary Surfactant-Associated Protein C/pharmacology , Pulmonary Surfactants/pharmacology , Sheep , Tissue Distribution
19.
Clin Sci (Lond) ; 133(21): 2159-2169, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31693732

ABSTRACT

Lung cancer ranks first in both incidence and mortality and is a major health concern worldwide. Upon recognition of specific antigens on tumor cells, complement-dependent cytotoxicity (CDC) is activated, arresting cell growth or inducing apoptosis. However, by overexpressing CD59, a membrane complement regulatory protein (mCRP), lung cancer cells develop resistance to CDC. We previously showed that virus-like particles (VLPs) of human JC polyomavirus (JCPyV) could be used as a gene therapy vector to carry a suicide gene expression plasmid with a lung-specific promoter (SP-B (surfactant protein B)) for lung adenocarcinomas. Herein, we designed a CD59-specific short hairpin RNA (shRNA) expression plasmid driven by SP-B (pSPB-shCD59) to effectively and specifically inhibit CD59 overexpression in lung cancer cells. Treatment of lung cancer cells in vitro with JCPyV VLPs containing pSPB-shCD59 (pSPB-shCD59/VLPs) induces CDC and death of cancer cells. Mice that were subcutaneously injected with human lung cancer cells showed an 87% inhibition in tumor growth after tail vein injection of pSPB-shCD59/VLPs. Moreover, in a mouse model of lung cancer metastasis, a reduction in the lung weight by 39%, compared with the control group, was observed in mice treated with pSPB-shCD59/VLPs after tail vein injection of human lung cancer cells. Furthermore, tissue sectioning showed that the number and size of tumors produced was significantly reduced in the lungs of mice in the treatment group than those of the untreated group, indicating inhibition of metastasis by pSPB-shCD59/VLPs. Together, these results demonstrate the potential of pSPB-shCD59/VLPs as a therapeutic agent for CD59 overexpressed lung cancer.


Subject(s)
Adenocarcinoma of Lung/therapy , CD59 Antigens/antagonists & inhibitors , Genetic Therapy/methods , Genetic Vectors/chemical synthesis , Lung Neoplasms/prevention & control , A549 Cells , Adenocarcinoma of Lung/secondary , Animals , Genetic Vectors/pharmacology , Humans , JC Virus , Lung Neoplasms/secondary , Male , Mice , Plasmids/chemical synthesis , Plasmids/pharmacology , Promoter Regions, Genetic , Pulmonary Surfactant-Associated Protein B/genetics , RNA, Small Interfering/pharmacology , Random Allocation , Xenograft Model Antitumor Assays
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL