Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 219
Filter
1.
PLoS One ; 19(3): e0297889, 2024.
Article in English | MEDLINE | ID: mdl-38483982

ABSTRACT

OBJECTIVE: Surfactant-specific proteins (SP) are responsible for the functional and structural integrity as well as for the stabilization of the intra-alveolar surfactant. Morphological lung maturation starts in rat lungs after birth. The aim of this study was to investigate whether the expression of the hydrophilic SP-A and the hydrophobic SP-B is associated with characteristic postnatal changes characterizing morphological lung maturation. METHODS: Stereological methods were performed on the light microscope. Using immunohistochemical and molecular biological methods (Western Blot, RT-qPCR), the SP-A and SP-B of adult rat lungs and of those with different postnatal developmental stages (3, 7, 14 and 21 days after birth) were characterized. RESULTS: As signs of alveolarization the total septal surface and volume increased and the septal thickness decreased. The significantly highest relative surface fraction of SP-A labeled alveolar epithelial cells type II (AEII) was found together with the highest relative SP-A gene expression before the alveolarization (3th postnatal day). With the downregulation of SP-A gene expression during and after alveolarization (between postnatal days 7 and 14), the surface fraction of the SP-A labeled AEII also decreased, so they are lowest in adult animals. The surface fraction of SP-B labeled AEII and the SP-B gene expression showed the significantly highest levels in adults, the protein expression increased also significantly at the end of morphological lung maturation. There were no alterations in the SP-B expression before and during alveolarization until postnatal day 14. The protein expression as well as the gene expression of SP-A and SP-B correlated very well with the total surface of alveolar septa independent of the postnatal age. CONCLUSION: The expression of SP-A and SP-B is differentially associated with morphological lung maturation and correlates with increased septation of alveoli as indirect clue for alveolarization.


Subject(s)
Pulmonary Surfactants , Surface-Active Agents , Rats , Animals , Surface-Active Agents/metabolism , Pulmonary Surfactants/metabolism , Lung/metabolism , Pulmonary Alveoli , Pulmonary Surfactant-Associated Proteins/genetics , Pulmonary Surfactant-Associated Proteins/metabolism , Lipoproteins/metabolism
2.
PLoS One ; 17(4): e0267155, 2022.
Article in English | MEDLINE | ID: mdl-35476695

ABSTRACT

Surfactant protein C (SP-C) has several functions in pulmonary surfactant. These include the transfer of lipids between different membrane structures, a role in surfactant recycling and homeostasis, and involvement in modulation of the innate defense system. Despite these important functions, the structures of functional SP-C complexes have remained unclear. SP-C is known to exist as a primarily α-helical structure with an apparently unstructured N-terminal region, yet there is recent evidence that the functions of SP-C could be associated with the formation of SP-C dimers and higher oligomers. In this work, we used molecular dynamics simulations, two-dimensional umbrella sampling, and well-tempered metadynamics to study the details of SP-C dimerization. The results suggest that SP-C dimerizes in pulmonary surfactant membranes, forming dimers of different topologies. The simulations identified a dimerization motif region V21xxxVxxxGxxxM33 that is much larger than the putative A30xxxG34 motif that is commonly assumed to control the dimerization of some α-helical transmembrane domains. The results provide a stronger basis for elucidating how SP-C functions in concert with other surfactant proteins.


Subject(s)
Pulmonary Surfactant-Associated Protein C , Pulmonary Surfactants , Dimerization , Pulmonary Surfactant-Associated Protein C/metabolism , Pulmonary Surfactant-Associated Proteins/metabolism , Pulmonary Surfactants/metabolism , Surface-Active Agents
3.
Balkan Med J ; 39(1): 130-139, 2022 03 14.
Article in English | MEDLINE | ID: mdl-35330560

ABSTRACT

Background: Acute respiratory distress syndrome (ARDS) is a severe disorder that is related to a high mortality. Mesenchymal stem cells (MSCs) have shown strong effects in relieving lung injury. Aims: To determine the role of umbilical cord-derived MSCs (UC-MSCs) together with surfactant protein B (SP-B) in ARDS. Study Design: Animal experimentation. Methods: Immunophenotypic characteristics of UC-MSCs were identified. BALB/c mice were intratracheally administrated with lipopolysaccharide (LPS) and received UC-MSCs or UC-MSCs transfected with SP-B (UC-MSCs-SP-B). Pathological changes and lung injury degrees after transplantation were assessed by histological and biochemical analyses. Inflammatory chemokine and cytokine production in the bronchoalveolar lavage fluid (BALF) was measured using enzyme-linked immunoassay. Flow cytometry was used to examine macrophage phenotypes and differentiation of T-helper 17 (Th17) and T-regulatory (Treg) in the BALF. Results: Our results showed that isolated UC-MSCs possessed multilineage differentiation potential. SP-B transfection into UC-MSCs strengthened the effects of UC-MSCs on lung function repair in LPS-induced ARDS. UC-MSCs and UC-MSCs-SP-B attenuated cellular infiltration. Additionally, UC-MSCs and UC-MSCs-SP-B inhibited the inflammatory response by promoting M2-like polarization, as well as reduced Th17 differentiation and promoted Treg differentiation. Conclusion: UC-MSCs in combination with SP-B, alleviates inflammatory reaction in ARDS by regulating macrophage polarization.


Subject(s)
Lung Injury , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Pulmonary Surfactant-Associated Proteins/metabolism , Respiratory Distress Syndrome , Animals , Humans , Lipopolysaccharides/metabolism , Lung Injury/metabolism , Macrophages , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/metabolism , Mice , Receptors, Fc , Respiratory Distress Syndrome/therapy , Surface-Active Agents/metabolism , Umbilical Cord
4.
Cells ; 11(2)2022 01 11.
Article in English | MEDLINE | ID: mdl-35053350

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a progressive and fatal degenerative lung disease of unknown etiology. Although in its final stages it implicates, in a reactive manner, all lung cell types, the initial damage involves the alveolar epithelial compartment, in particular the alveolar epithelial type 2 cells (AEC2s). AEC2s serve dual progenitor and surfactant secreting functions, both of which are deeply impacted in IPF. Thus, we hypothesize that the size of the surfactant processing compartment, as measured by LysoTracker incorporation, allows the identification of different epithelial states in the IPF lung. Flow cytometry analysis of epithelial LysoTracker incorporation delineates two populations (Lysohigh and Lysolow) of AEC2s that behave in a compensatory manner during bleomycin injury and in the donor/IPF lung. Employing flow cytometry and transcriptomic analysis of cells isolated from donor and IPF lungs, we demonstrate that the Lysohigh population expresses all classical AEC2 markers and is drastically diminished in IPF. The Lysolow population, which is increased in proportion in IPF, co-expressed AEC2 and basal cell markers, resembling the phenotype of the previously identified intermediate AEC2 population in the IPF lung. In that regard, we provide an in-depth flow-cytometry characterization of LysoTracker uptake, HTII-280, proSP-C, mature SP-B, NGFR, KRT5, and CD24 expression in human lung epithelial cells. Combining functional analysis with extracellular and intracellular marker expression and transcriptomic analysis, we advance the current understanding of epithelial cell behavior and fate in lung fibrosis.


Subject(s)
Alveolar Epithelial Cells/metabolism , Amines/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Animals , Biomarkers/metabolism , Bleomycin , CD24 Antigen/metabolism , Epithelium/pathology , Gene Expression Profiling , Humans , Idiopathic Pulmonary Fibrosis/genetics , Keratin-5/metabolism , Mice, Inbred C57BL , Pulmonary Surfactant-Associated Proteins/metabolism , Receptors, Nerve Growth Factor/metabolism , Tissue Donors , Transcription, Genetic , Up-Regulation
5.
Physiol Res ; 70(S2): S195-S208, 2021 12 16.
Article in English | MEDLINE | ID: mdl-34913352

ABSTRACT

In this review, we discuss the role of pulmonary surfactant in the host defense against respiratory pathogens, including novel coronavirus SARS-CoV-2. In the lower respiratory system, the virus uses angiotensin-converting enzyme 2 (ACE2) receptor in conjunction with serine protease TMPRSS2, expressed by alveolar type II (ATII) cells as one of the SARS-CoV-2 target cells, to enter. ATII cells are the main source of surfactant. After their infection and the resulting damage, the consequences may be severe and may include injury to the alveolar-capillary barrier, lung edema, inflammation, ineffective gas exchange, impaired lung mechanics and reduced oxygenation, which resembles acute respiratory distress syndrome (ARDS) of other etiology. The aim of this review is to highlight the key role of ATII cells and reduced surfactant in the pathogenesis of the respiratory form of COVID-19 and to emphasize the rational basis for exogenous surfactant therapy in COVID-19 ARDS patients.


Subject(s)
Alveolar Epithelial Cells/metabolism , COVID-19/metabolism , Lung/metabolism , Pulmonary Surfactant-Associated Proteins/metabolism , SARS-CoV-2/pathogenicity , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/immunology , Alveolar Epithelial Cells/virology , Angiotensin-Converting Enzyme 2/metabolism , Animals , COVID-19/immunology , COVID-19/virology , Host-Pathogen Interactions , Humans , Lung/drug effects , Lung/immunology , Lung/virology , Pulmonary Surfactants/therapeutic use , Receptors, Virus/metabolism , SARS-CoV-2/immunology , Serine Endopeptidases/metabolism , Virus Internalization , COVID-19 Drug Treatment
6.
Eur Rev Med Pharmacol Sci ; 25(13): 4639-4643, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34286505

ABSTRACT

OBJECTIVE: Acute respiratory distress syndrome (ARDS) is characterized by quantitative and qualitative changes in surfactant composition, leading to surfactant dysregulation with alveolar collapse and acute respiratory hypoxic failure. Recently, surfactant has been hypothesized to play a relevant role in COVID-19, representing a strong defender against SARS-CoV-2 infection. The aim of our work was the study of immunohistochemical surfactant expression in the lungs of patients died following SARS-CoV-2 ARDS, in order to shed light on a possible therapeutic surfactant administration. PATIENTS AND METHODS: We investigated four patients who died due to ARDS following SARS-COV-2 infection and four patients submitted to lung biopsy, in the absence of SARS-CoV-2 infection. In all 8 cases, lung specimens were immunostained with anti-surfactant protein A (SP-A) and B (SP-B). RESULTS: In control subjects, reactivity for SP-B was restricted to type II alveolar cells. Immunostaining for SP-A was observed on the surface of alveolar spaces. In the COVID-19 positive lungs, immunoreactivity for SP-B was similar to that observed in control lungs; SP-A was strongly expressed along the alveolar wall. Moreover, dense aggregates of SP-A positive material were observed in the alveolar spaces. CONCLUSIONS: Our immunohistochemical data show the dysregulation of surfactant production in COVID-19 patients, particularly regarding SP-A expression. The increased presence of SP-A in condensed masses inside alveolar spaces could invalidate the therapeutic efficacy of the treatment with exogenous surfactant.


Subject(s)
COVID-19/metabolism , Immunohistochemistry , Protein Precursors/analysis , Pulmonary Surfactant-Associated Protein A/analysis , Pulmonary Surfactant-Associated Proteins/analysis , COVID-19/diagnostic imaging , Humans , Protein Precursors/genetics , Protein Precursors/metabolism , Pulmonary Alveoli/diagnostic imaging , Pulmonary Alveoli/metabolism , Pulmonary Surfactant-Associated Protein A/genetics , Pulmonary Surfactant-Associated Protein A/metabolism , Pulmonary Surfactant-Associated Proteins/genetics , Pulmonary Surfactant-Associated Proteins/metabolism , Retrospective Studies , SARS-CoV-2/isolation & purification , SARS-CoV-2/metabolism
7.
Am J Respir Cell Mol Biol ; 65(4): 442-460, 2021 10.
Article in English | MEDLINE | ID: mdl-34101541

ABSTRACT

Alveolar epithelial type 2 cells (AEC2s), the facultative progenitors of lung alveoli, are typically identified through the use of the canonical markers, SFTPC and ABCA3. Self-renewing AEC2-like cells have been generated from human induced pluripotent stem cells (iPSCs) through the use of knock-in SFTPC fluorochrome reporters. However, developmentally, SFTPC expression onset begins in the fetal distal lung bud tip and thus is not specific to mature AEC2s. Furthermore, SFTPC reporters appear to identify only those iPSC-derived AEC2s (iAEC2s) expressing the highest SFTPC levels. Here, we generate an ABCA3 knock-in GFP fusion reporter (ABCA3:GFP) that enables the purification of iAEC2s while allowing visualization of lamellar bodies, organelles associated with AEC2 maturation. Using an SFTPCtdTomato and ABCA3:GFP bifluorescent line for in vitro distal lung-directed differentiation, we observe later onset of ABCA3:GFP expression and broader identification of the subsequently emerging iAEC2 population based on ABCA3:GFP expression compared with SFTPCtdTomato expression. Comparing ABCA3:GFP/SFTPCtdTomato double-positive with ABCA3:GFP single-positive (SP) cells by RNA sequencing and functional studies reveals iAEC2 cellular heterogeneity with both populations functionally processing surfactant proteins but the SP cells exhibiting faster growth kinetics, increased clonogenicity, increased expression of progenitor markers, lower levels of SFTPC expression, and lower levels of AEC2 maturation markers. Over time, we observe that each population (double-positive and SP) gives rise to the other and each can serve as the parents of indefinitely self-renewing iAEC2 progeny. Our results indicate that iAEC2s are a heterogeneous population of cells with differing proliferation versus maturation properties, the majority of which can be tracked and purified using the ABCA3:GFP reporter or surrogate cell surface proteins, such as SLC34A2 and CPM.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Alveolar Epithelial Cells/cytology , Induced Pluripotent Stem Cells/cytology , Pulmonary Alveoli/cytology , Pulmonary Surfactant-Associated Protein C/metabolism , Cell Differentiation/physiology , Epithelial Cells/metabolism , Humans , Lung/metabolism , Pulmonary Surfactant-Associated Proteins/metabolism
8.
Int J Mol Sci ; 22(5)2021 Mar 04.
Article in English | MEDLINE | ID: mdl-33806395

ABSTRACT

Alveolar type II (ATII) cells are a key structure of the distal lung epithelium, where they exert their innate immune response and serve as progenitors of alveolar type I (ATI) cells, contributing to alveolar epithelial repair and regeneration. In the healthy lung, ATII cells coordinate the host defense mechanisms, not only generating a restrictive alveolar epithelial barrier, but also orchestrating host defense mechanisms and secreting surfactant proteins, which are important in lung protection against pathogen exposure. Moreover, surfactant proteins help to maintain homeostasis in the distal lung and reduce surface tension at the pulmonary air-liquid interface, thereby preventing atelectasis and reducing the work of breathing. ATII cells may also contribute to the fibroproliferative reaction by secreting growth factors and proinflammatory molecules after damage. Indeed, various acute and chronic diseases are associated with intensive inflammation. These include oedema, acute respiratory distress syndrome, fibrosis and numerous interstitial lung diseases, and are characterized by hyperplastic ATII cells which are considered an essential part of the epithelialization process and, consequently, wound healing. The aim of this review is that of revising the physiologic and pathologic role ATII cells play in pulmonary diseases, as, despite what has been learnt in the last few decades of research, the origin, phenotypic regulation and crosstalk of these cells still remain, in part, a mystery.


Subject(s)
Alveolar Epithelial Cells/pathology , Alveolar Epithelial Cells/physiology , Lung Diseases/physiopathology , Lung/physiology , Alveolar Epithelial Cells/cytology , Animals , COVID-19/physiopathology , Humans , Immunity, Innate , Ions/metabolism , Lung/anatomy & histology , Lung Diseases/etiology , Lung Diseases/pathology , Pulmonary Surfactant-Associated Proteins/metabolism , Regeneration
9.
Toxicol Appl Pharmacol ; 417: 115470, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33647319

ABSTRACT

Bleomycin is a cancer therapeutic known to cause lung injury which progresses to fibrosis. Evidence suggests that macrophages contribute to this pathological response. Tumor necrosis factor (TNF)α is a macrophage-derived pro-inflammatory cytokine implicated in lung injury. Herein, we investigated the role of TNFα in macrophage responses to bleomycin. Treatment of mice with bleomycin (3 U/kg, i.t.) caused histopathological changes in the lung within 3 d which culminated in fibrosis at 21 d. This was accompanied by an early (3-7 d) influx of CD11b+ and iNOS+ macrophages into the lung, and Arg-1+ macrophages at 21 d. At this time, epithelial cell dysfunction, defined by increases in total phospholipids and SP-B was evident. Treatment of mice with anti-TNFα antibody (7.5 mg/kg, i.v.) beginning 15-30 min after bleomycin, and every 5 d thereafter reduced the number and size of fibrotic foci and restored epithelial cell function. Flow cytometric analysis of F4/80+ alveolar macrophages (AM) isolated by bronchoalveolar lavage and interstitial macrophages (IM) by tissue digestion identified resident (CD11b-CD11c+) and immature infiltrating (CD11b+CD11c-) AM, and mature (CD11b+CD11c+) and immature (CD11b+CD11c-) IM subsets in bleomycin treated mice. Greater numbers of mature (CD11c+) infiltrating (CD11b+) AM expressing the anti-inflammatory marker, mannose receptor (CD206) were observed at 21 d when compared to 7 d post bleomycin. Mature proinflammatory (Ly6C+) IM were greater at 7 d relative to 21 d. These cells transitioned into mature anti-inflammatory/pro-fibrotic (CD206+) IM between 7 and 21 d. Anti-TNFα antibody heightened the number of CD11b+ AM in the lung without altering their activation state. Conversely, it reduced the abundance of mature proinflammatory (Ly6C+) IM in the tissue at 7 d and immature pro-fibrotic IM at 21 d. Taken together, these data suggest that TNFα inhibition has beneficial effects in bleomycin induced injury, restoring epithelial function and reducing numbers of profibrotic IM and the extent of pulmonary fibrosis.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Lung/drug effects , Macrophage Activation/drug effects , Macrophages/drug effects , Pneumonia/prevention & control , Pulmonary Fibrosis/prevention & control , Tumor Necrosis Factor Inhibitors/pharmacology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Animals , Bleomycin , Disease Models, Animal , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Fibrosis , Lung/metabolism , Lung/pathology , Macrophages/metabolism , Macrophages/pathology , Male , Mice, Inbred C57BL , Phenotype , Phospholipids/metabolism , Pneumonia/chemically induced , Pneumonia/metabolism , Pneumonia/pathology , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Pulmonary Surfactant-Associated Proteins/metabolism , Tumor Necrosis Factor-alpha/metabolism
10.
Am J Physiol Lung Cell Mol Physiol ; 320(5): L942-L957, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33719563

ABSTRACT

The ability of pulmonary surfactant to reduce alveolar surface tension requires adequate levels of surfactant protein B (SP-B). Dexamethasone (DEX) increases human SP-B expression, in part, through increased SP-B mRNA stability. A 30-nt-long hairpin element (RBE) in the 3'-untranslated region of human SP-B mRNA mediates both DEX-induced and intrinsic mRNA stabilities, but the mechanism is unknown. Proteomic analysis of RBE-interacting proteins identified a primate-specific protein, RNA-binding motif X-linked-like-3 (RBMXL3). siRNA directed against RBMXL3 reduces DEX-induced SP-B mRNA expression in human bronchoalveolar cells. Human SP-B mRNA stability, measured by our dual cistronic plasmid assay, is unaffected by DEX in mouse lung epithelial cells lacking RBMXL3, but DEX increases human SP-B mRNA stability when RBMXL3 is expressed and requires the RBE. In the absence of DEX, RBE interacts with cellular proteins, reducing intrinsic SP-B mRNA stability in human and mouse lung epithelial cells. RBMXL3 specifically binds the RBE in vitro, whereas RNA immunoprecipitation and affinity chromatography analyses indicate that binding is enhanced in the presence of DEX. These results describe a model where intrinsic stability of human SP-B mRNA is reduced through binding of cellular mRNA decay factors to RBE, which is then relieved through DEX-enhanced binding of primate-specific RBMXL3.


Subject(s)
Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Protein Precursors/metabolism , Pulmonary Surfactant-Associated Proteins/metabolism , RNA Stability/drug effects , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , A549 Cells , Animals , HEK293 Cells , Humans , Mice , Protein Precursors/genetics , Pulmonary Surfactant-Associated Proteins/genetics , RNA, Messenger/genetics , RNA-Binding Proteins/genetics
11.
Sci Rep ; 11(1): 728, 2021 01 12.
Article in English | MEDLINE | ID: mdl-33436647

ABSTRACT

Therapeutic hypothermia (TH) enhances pulmonary surfactant performance in vivo by molecular mechanisms still unknown. Here, the interfacial structure and the composition of lung surfactant films have been analysed in vitro under TH as well as the molecular basis of its improved performance both under physiological and inhibitory conditions. The biophysical activity of a purified porcine surfactant was tested under slow and breathing-like dynamics by constrained drop surfactometry (CDS) and in the captive bubble surfactometer (CBS) at both 33 and 37 °C. Additionally, the temperature-dependent surfactant activity was also analysed upon inhibition by plasma and subsequent restoration by further surfactant supplementation. Interfacial performance was correlated with lateral structure and lipid composition of films made of native surfactant. Lipid/protein mixtures designed as models to mimic different surfactant contexts were also studied. The capability of surfactant to drastically reduce surface tension was enhanced at 33 °C. Larger DPPC-enriched domains and lower percentages of less active lipids were detected in surfactant films exposed to TH-like conditions. Surfactant resistance to plasma inhibition was boosted and restoration therapies were more effective at 33 °C. This may explain the improved respiratory outcomes observed in cooled patients with acute respiratory distress syndrome and opens new opportunities in the treatment of acute lung injury.


Subject(s)
Hypothermia, Induced/methods , Lung/physiology , Phospholipids/metabolism , Pulmonary Surfactant-Associated Proteins/metabolism , Pulmonary Surfactants/metabolism , Respiratory Physiological Phenomena , Animals , Biophysics , Phase Transition , Pulmonary Surfactants/chemistry , Swine
12.
Commun Biol ; 4(1): 137, 2021 01 29.
Article in English | MEDLINE | ID: mdl-33514845

ABSTRACT

Lamellar bodies (LBs) are surfactant-rich organelles in alveolar cells. LBs disassemble into a lipid-protein network that reduces surface tension and facilitates gas exchange in the alveolar cavity. Current knowledge of LB architecture is predominantly based on electron microscopy studies using disruptive sample preparation methods. We established and validated a post-correlation on-lamella cryo-correlative light and electron microscopy approach for cryo-FIB milled cells to structurally characterize and validate the identity of LBs in their unperturbed state. Using deconvolution and 3D image registration, we were able to identify fluorescently labeled membrane structures analyzed by cryo-electron tomography. In situ cryo-electron tomography of A549 cells as well as primary Human Small Airway Epithelial Cells revealed that LBs are composed of membrane sheets frequently attached to the limiting membrane through "T"-junctions. We report a so far undescribed outer membrane dome protein complex (OMDP) on the limiting membrane of LBs. Our data suggest that LB biogenesis is driven by parallel membrane sheet import and by the curvature of the limiting membrane to maximize lipid storage capacity.


Subject(s)
Cryoelectron Microscopy , Electron Microscope Tomography , Image Interpretation, Computer-Assisted , Imaging, Three-Dimensional , Intracellular Membranes/ultrastructure , Lung Neoplasms/ultrastructure , Organelles/ultrastructure , Pulmonary Alveoli/ultrastructure , A549 Cells , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Organelles/metabolism , Pulmonary Alveoli/metabolism , Pulmonary Surfactant-Associated Proteins/metabolism , Recombinant Fusion Proteins/metabolism
13.
Am J Respir Cell Mol Biol ; 64(4): 504-514, 2021 04.
Article in English | MEDLINE | ID: mdl-33493427

ABSTRACT

Alveolar epithelial type II (AT2) cells secrete pulmonary surfactant via lamellar bodies (LBs). Abnormalities in LBs are associated with pulmonary disorders, including fibrosis. However, high-content screening (HCS) for LB abnormalities is limited by the lack of understanding of AT2 cell functions. In the present study, we have developed LB cells harboring LB-like organelles that secrete surfactant proteins. These cells were more similar to AT2 cells than to parental A549 cells. LB cells recapitulated amiodarone (AMD)-induced LB enlargement, similar to AT2 cells of patients exposed to AMD. To reverse AMD-induced LB abnormalities, we performed HCS of approved drugs and identified 2-hydroxypropyl-ß-cyclodextrin (HPßCD), a cyclic oligosaccharide, as a potential therapeutic agent. A transcriptome analysis revealed that HPßCD modulates lipid homeostasis. In addition, HPßCD inhibited AMD-induced LB abnormalities in human induced pluripotent stem cell-derived AT2 cells. Our results demonstrate that LB cells are useful for HCS and suggest that HPßCD is a candidate therapeutic agent for AMD-induced interstitial pneumonia.


Subject(s)
2-Hydroxypropyl-beta-cyclodextrin/pharmacology , Alveolar Epithelial Cells/drug effects , Amiodarone/toxicity , Induced Pluripotent Stem Cells/drug effects , Lipid Metabolism/drug effects , A549 Cells , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/pathology , High-Throughput Screening Assays , Homeostasis , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Protein Precursors/metabolism , Pulmonary Surfactant-Associated Protein C/metabolism , Pulmonary Surfactant-Associated Protein D/metabolism , Pulmonary Surfactant-Associated Proteins/metabolism
14.
Protein Expr Purif ; 179: 105801, 2021 03.
Article in English | MEDLINE | ID: mdl-33248225

ABSTRACT

While the discovery of antibiotics has made a huge contribution to medicine, bacteria that are resistant to many antibiotics pose new challenges to medicine. Antimicrobial peptides (AMPs), a new kind of antibiotics, have attracted people's attention because they are not prone to drug resistance. In this study, glutathione transferase (GST) was used as a fusion partner to recombinantly expressed rat lung surfactant protein B precursor (proSP-B) in E. coli pLySs. Cck-8 evaluated the cytotoxicity of the fusion protein and calculated its 50% inhibitory concentration (IC50). The purified peptides showed broad-spectrum antibacterial activity using filter paper method and MIC, and propidium iodide (PI) was used to explore the antibacterial mechanism against Staphylococcus aureus. In addition, the pEGFP-N2-proSP-B vector was constructed to explore the localization of proSP-B in CCL-149 cells. We found that proSP-B has obvious antibacterial activity against Gram-positive bacteria, Gram-negative bacteria and fungi, and has broad-spectrum antibacterial activity. Besides, proSP-B fusion protein has low toxicity and can change the permeability of Staphylococcus aureus cell membrane to realize its antibacterial. For these reasons, proSP-B can be used as a potential natural antibacterial drug.


Subject(s)
Anti-Bacterial Agents , Pulmonary Surfactant-Associated Proteins , Recombinant Proteins , Animals , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Escherichia coli/genetics , Fungi/drug effects , Lung/chemistry , Microbial Sensitivity Tests , Pulmonary Surfactant-Associated Proteins/genetics , Pulmonary Surfactant-Associated Proteins/metabolism , Pulmonary Surfactant-Associated Proteins/pharmacology , RNA/isolation & purification , Rats , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology
15.
Eur J Histochem ; 64(s2)2020 Oct 01.
Article in English | MEDLINE | ID: mdl-33334090

ABSTRACT

Considerable evidence has verified that microRNAs (miRNAs) play important roles in various cellular processes including differentiation. However, the regulatory roles of miRNAs involved in the differentiation of induced pluripotent stem cells (iPSC) into lung epithelial cells are still unknown. In this study, we first evaluated the current protocols to differentiate iPSC into alveolar epithelial type II (AEC II) cells, but the efficiency is low. We next identified that miR-22 can efficiently enhance the differentiation of iPSC into AEC II cells under the stimulation of proper growth factors and growing on appropriate matrix. Moreover, the AEC II cells generated from iPSC with miR-22 overexpression can proliferate and secrete lung surfactant. Here, we discovered a previously unknown interaction between miR-22 and iPSC differentiation but also provide a potential target for the effective derivation of AEC II from iPSCs for cell-based therapy.


Subject(s)
Alveolar Epithelial Cells/metabolism , Cell Differentiation/physiology , Induced Pluripotent Stem Cells/metabolism , MicroRNAs/metabolism , Animals , HEK293 Cells , Humans , Male , Mice, Inbred C57BL , Pulmonary Surfactant-Associated Proteins/metabolism
16.
Elife ; 92020 11 24.
Article in English | MEDLINE | ID: mdl-33228849

ABSTRACT

We establish a murine lung-on-chip infection model and use time-lapse imaging to reveal the dynamics of host-Mycobacterium tuberculosis interactions at an air-liquid interface with a spatiotemporal resolution unattainable in animal models and to probe the direct role of pulmonary surfactant in early infection. Surfactant deficiency results in rapid and uncontrolled bacterial growth in both macrophages and alveolar epithelial cells. In contrast, under normal surfactant levels, a significant fraction of intracellular bacteria are non-growing. The surfactant-deficient phenotype is rescued by exogenous addition of surfactant replacement formulations, which have no effect on bacterial viability in the absence of host cells. Surfactant partially removes virulence-associated lipids and proteins from the bacterial cell surface. Consistent with this mechanism, the attenuation of bacteria lacking the ESX-1 secretion system is independent of surfactant levels. These findings may partly explain why smokers and elderly persons with compromised surfactant function are at increased risk of developing active tuberculosis.


Tuberculosis is a contagious respiratory disease caused by the bacterium Mycobacterium tuberculosis. Droplets in the air carry these bacteria deep into the lungs, where they cling onto and infect lung cells. Only small droplets, holding one or two bacteria, can reach the right cells, which means that just a couple of bacterial cells can trigger an infection. But people respond differently to the bacteria: some develop active and fatal forms of tuberculosis, while many show no signs of infection. With no effective tuberculosis vaccine for adults, understanding why individuals respond differently to Mycobacterium tuberculosis may help develop treatments. Different responses to Mycobacterium tuberculosis may stem from the earliest stages of infection, but these stages are difficult to study. For one thing, tracking the movements of the few bacterial cells that initiate infection is tricky. For another, studying the molecules, called 'surfactants', that the lungs produce to protect themselves from tuberculosis can prove difficult because these molecules are necessary for the lungs to inflate and deflate normally. Normally, the role of a molecule can be studied by genetically modifying an animal so it does not produce the molecule in question, which provides information as to its potential roles. Unfortunately, due to the role of surfactants in normal breathing, animals lacking them die. Therefore, to reveal the role of some of surfactants in tuberculosis, Thacker et al. used 'lung-on-chip' technology. The 'chip' (a transparent device made of a polymer compatible with biological tissues) is coated with layers of cells and has channels to simulate air and blood flow. To see what effects surfactants have on M. tuberculosis bacteria, Thacker et al. altered the levels of surfactants produced by the cells on the lung-on-chip device. Two types of mouse cells were grown on the chip: lung cells and immune cells. When cells lacked surfactants, bacteria grew rapidly on both lung and immune cells, but when surfactants were present bacteria grew much slower on both cell types, or did not grow at all. Further probing showed that the surfactants pulled out proteins and fats on the surface of M. tuberculosis that help the bacteria to infect their host, highlighting the protective role of surfactants in tuberculosis. These findings lay the foundations for a system to study respiratory infections without using animals. This will allow scientists to study the early stages of Mycobacterium tuberculosis infection, which is crucial for finding ways to manage tuberculosis.


Subject(s)
Alveolar Epithelial Cells/microbiology , Lab-On-A-Chip Devices , Microfluidic Analytical Techniques/instrumentation , Mycobacterium tuberculosis/growth & development , Pulmonary Surfactant-Associated Proteins/metabolism , Tuberculosis, Pulmonary/microbiology , Alveolar Epithelial Cells/metabolism , Animals , Bacterial Load , Bacterial Proteins/genetics , Cells, Cultured , Disease Models, Animal , Female , Host-Pathogen Interactions , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/microbiology , Mice, Inbred C57BL , Mice, Transgenic , Microbial Viability , Microscopy, Video , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/pathogenicity , Pulmonary Surfactant-Associated Proteins/genetics , Time Factors , Time-Lapse Imaging , Tuberculosis, Pulmonary/genetics , Tuberculosis, Pulmonary/metabolism , Virulence
17.
Mol Cell Proteomics ; 19(9): 1523-1532, 2020 09.
Article in English | MEDLINE | ID: mdl-32581039

ABSTRACT

Communication between individuals via molecules, termed chemosignaling, is widespread among animal and plant species. However, we lack knowledge on the specific functions of the substances involved for most systems. The femoral gland is an organ that secretes a waxy substance involved in chemical communication in lizards. Although the lipids and volatile substances secreted by the femoral glands have been investigated in several biochemical studies, the protein composition and functions of secretions remain completely unknown. Applying a proteomic approach, we provide the first attempt to comprehensively characterize the protein composition of femoral gland secretions from the Galápagos marine iguana. Using samples from several organs, the marine iguana proteome was assembled by next-generation sequencing and MS, resulting in 7513 proteins. Of these, 4305 proteins were present in the femoral gland, including keratins, small serum proteins, and fatty acid-binding proteins. Surprisingly, no proteins with discernible roles in partner recognition or inter-species communication could be identified. However, we did find several proteins with direct associations to the innate immune system, including lysozyme C, antileukoproteinase (ALP), pulmonary surfactant protein (SFTPD), and galectin (LGALS1) suggesting that the femoral glands function as an important barrier to infection. Furthermore, we report several novel anti-microbial peptides from the femoral glands that show similar action against Escherichia coli and Bacillus subtilis such as oncocin, a peptide known for its effectiveness against Gram-negative pathogens. This proteomics data set is a valuable resource for future functional protein analysis and demonstrates that femoral gland secretions also perform functions of the innate immune system.


Subject(s)
Anti-Infective Agents/metabolism , Anti-Infective Agents/pharmacology , Iguanas/metabolism , Immune System/metabolism , Immunity, Innate , Proteome/metabolism , Transcriptome , Animals , Apoproteins/genetics , Apoproteins/metabolism , Bacillus subtilis/drug effects , Brain/metabolism , Chemotactic Factors/genetics , Chemotactic Factors/metabolism , Ecuador , Endopeptidases/genetics , Endopeptidases/metabolism , Escherichia coli/drug effects , Galectins/genetics , Galectins/metabolism , Heart/physiology , High-Throughput Nucleotide Sequencing , Humans , Iguanas/genetics , Iguanas/immunology , Immunity, Innate/genetics , Lung/metabolism , Muramidase/genetics , Muramidase/metabolism , Muscles/metabolism , Myocardium/metabolism , Organ Specificity , Proteome/genetics , Proteome/immunology , Proteomics , Pulmonary Surfactant-Associated Proteins/genetics , Pulmonary Surfactant-Associated Proteins/metabolism , Skin/metabolism , Tandem Mass Spectrometry , Transcriptome/genetics
18.
Int J Mol Sci ; 21(10)2020 May 25.
Article in English | MEDLINE | ID: mdl-32466119

ABSTRACT

Pulmonary surfactant is a lipid/protein complex synthesized by the alveolar epithelium and secreted into the airspaces, where it coats and protects the large respiratory air-liquid interface. Surfactant, assembled as a complex network of membranous structures, integrates elements in charge of reducing surface tension to a minimum along the breathing cycle, thus maintaining a large surface open to gas exchange and also protecting the lung and the body from the entrance of a myriad of potentially pathogenic entities. Different molecules in the surfactant establish a multivalent crosstalk with the epithelium, the immune system and the lung microbiota, constituting a crucial platform to sustain homeostasis, under health and disease. This review summarizes some of the most important molecules and interactions within lung surfactant and how multiple lipid-protein and protein-protein interactions contribute to the proper maintenance of an operative respiratory surface.


Subject(s)
Alveolar Epithelial Cells/metabolism , Homeostasis , Pulmonary Surfactant-Associated Proteins/metabolism , Alveolar Epithelial Cells/microbiology , Alveolar Epithelial Cells/physiology , Animals , Humans , Lipid Metabolism
19.
J Cell Mol Med ; 24(12): 7067-7071, 2020 06.
Article in English | MEDLINE | ID: mdl-32363643

ABSTRACT

Bronchopulmonary dysplasia (BPD) is a major cause of mortality and morbidity in premature infants, characterized by alveolar simplification, surfactant deficiency, and respiratory distress. In the present study, we have investigated the functional roles of sumoylated CCAAT/enhancer binding protein alpha (C/EBPα) in the BPD rat model. A significant increase in small ubiquitin-like modifier 1 (SUMO1) and sumoylated C/EBPα protein levels were observed in BPD rats, and the levels of the sumoylated C/EBPα were associated with the pulmonary surfactant proteins (SPs). In order to confirm the role of sumoylated C/EBPα in BPD rats, SUMO1 was knocked down by lentiviral transfection of neonatal rat lungs with SUMO1-RNAi-LV. We found that the expression of C/EBPα and surfactant proteins increased following SUMO1 knockdown. Furthermore, the relatively low decrease in the levels of C/EBPα sumoylation was correlated with reduced glycogen consumption. Besides, co-immunoprecipitation assays revealed that sumoylation is involved in the regulation of the interaction between C/EBPα and TGFß2 in the lung. In conclusion, our findings indicate that sumoylation may act as a negative regulator of the C/EBPα-mediated transactivation in BPD rats.


Subject(s)
Bronchopulmonary Dysplasia/pathology , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Cell Differentiation , Lung/pathology , Sumoylation , Animals , Disease Models, Animal , Gene Knockdown Techniques , Glycogen/metabolism , Protein Binding , Pulmonary Surfactant-Associated Proteins/metabolism , Rats, Sprague-Dawley , SUMO-1 Protein/metabolism , Transforming Growth Factor beta2/metabolism
20.
Reprod Toxicol ; 94: 40-47, 2020 06.
Article in English | MEDLINE | ID: mdl-32330513

ABSTRACT

Gestational vitamin D deficiency is associated with pulmonary diseases. This study aimed to investigate the effect of gestational vitamin D deficiency on fetal lung development in mice. Absolute and relative weights of fetal lungs were reduced in vitamin D deficient (VDD) group. Incrassate mesenchyme, measured by septal wall thickness, accompanied by lessened saccular space, was shown in VDD group. Numerous immature type II pneumocytes, as determined by PAS staining, were observed in VDD group. Moreover, increased Ki67-positive cells, a marker of cell proliferation, was detected in VDD group. The additional experiments showed that Sftpa, Sftpb, Sftpc and Sftpd, four surfactant genes, were downregulated and pro-surfactant protein B was reduced in VDD group. FoxA1, FoxA2 and TTF-1, three transcription factors that regulate surfactant genes, and VEGF, a key regulator for pulmonary maturation, were downregulated in VDD group. These results suggest that gestational vitamin D deficiency impairs fetal lung development partially through suppressing type II pneumocyte differentiation.


Subject(s)
Alveolar Epithelial Cells/cytology , Fetal Development , Lung/growth & development , Vitamin D Deficiency/complications , Alveolar Epithelial Cells/metabolism , Animals , Cell Differentiation , Cytokines/genetics , DNA-Binding Proteins/genetics , Female , Fetus , Hepatocyte Nuclear Factor 3-alpha/genetics , Hepatocyte Nuclear Factor 3-beta/genetics , Humans , Ki-67 Antigen/metabolism , Male , Mice , Pregnancy , Pulmonary Surfactant-Associated Proteins/genetics , Pulmonary Surfactant-Associated Proteins/metabolism , Transcription Factors/genetics , Vascular Endothelial Growth Factor A/genetics , Vitamin D/blood , Vitamin D Deficiency/blood , Vitamin D Deficiency/genetics , Vitamin D Deficiency/metabolism , Vitamins/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...