Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
Add more filters

Publication year range
1.
Am J Respir Crit Care Med ; 203(4): 471-483, 2021 02 15.
Article in English | MEDLINE | ID: mdl-32877203

ABSTRACT

Rationale: The regeneration and replacement of lung cells or tissues from induced pluripotent stem cell- or embryonic stem cell-derived cells represent future therapies for life-threatening pulmonary disorders but are limited by technical challenges to produce highly differentiated cells able to maintain lung function. Functional lung tissue-containing airways, alveoli, vasculature, and stroma have never been produced via directed differentiation of embryonic stem cells (ESCs) or induced pluripotent stem cells. We sought to produce all tissue components of the lung from bronchi to alveoli by embryo complementation.Objectives: To determine whether ESCs are capable of generating lung tissue in Nkx2-1-/- mouse embryos with lung agenesis.Methods: Blastocyst complementation was used to produce chimeras from normal mouse ESCs and Nkx2-1-/- embryos, which lack pulmonary tissues. Nkx2-1-/- chimeras were examined using immunostaining, transmission electronic microscopy, fluorescence-activated cell sorter analysis, and single-cell RNA sequencing.Measurements and Main Results: Although peripheral pulmonary and thyroid tissues are entirely lacking in Nkx2-1 gene-deleted embryos, pulmonary and thyroid structures in Nkx2-1-/- chimeras were restored after ESC complementation. Respiratory epithelial cell lineages in restored lungs of Nkx2-1-/- chimeras were derived almost entirely from ESCs, whereas endothelial, immune, and stromal cells were mosaic. ESC-derived cells from multiple respiratory cell lineages were highly differentiated and indistinguishable from endogenous cells based on morphology, ultrastructure, gene expression signatures, and cell surface proteins used to identify cell types by fluorescence-activated cell sorter.Conclusions: Lung and thyroid tissues were generated in vivo from ESCs by blastocyst complementation. Nkx2-1-/- chimeras can be used as "bioreactors" for in vivo differentiation and functional studies of ESC-derived progenitor cells.


Subject(s)
Blastocyst/physiology , Cell Differentiation/physiology , Embryonic Stem Cells/physiology , Lung Diseases/therapy , Lung/growth & development , Thyroid Gland/growth & development , Tissue Engineering/methods , Animals , Cell Differentiation/genetics , Humans , Mice , Models, Animal
2.
Int J Mol Sci ; 22(20)2021 Oct 15.
Article in English | MEDLINE | ID: mdl-34681806

ABSTRACT

As key components of innate immunity, lung antimicrobial proteins play a critical role in warding off invading respiratory pathogens. Lung surfactant protein A (SP-A) exerts synergistic antimicrobial activity with the N-terminal segment of the SP-B proprotein (SP-BN) against Klebsiella pneumoniae K2 in vivo. However, the factors that govern SP-A/SP-BN antimicrobial activity are still unclear. The aim of this study was to identify the mechanisms by which SP-A and SP-BN act synergistically against K. pneumoniae, which is resistant to either protein alone. The effect of these proteins on K. pneumoniae was studied by membrane permeabilization and depolarization assays and transmission electron microscopy. Their effects on model membranes of the outer and inner bacterial membranes were analyzed by differential scanning calorimetry and membrane leakage assays. Our results indicate that the SP-A/SP-BN complex alters the ultrastructure of K. pneumoniae by binding to lipopolysaccharide molecules present in the outer membrane, forming packing defects in the membrane that may favor the translocation of both proteins to the periplasmic space. The SP-A/SP-BN complex depolarized and permeabilized the inner membrane, perhaps through the induction of toroidal pores. We conclude that the synergistic antimicrobial activity of SP-A/SP-BN is based on the capability of this complex, but not either protein alone, to alter the integrity of bacterial membranes.


Subject(s)
Anti-Bacterial Agents/pharmacology , Klebsiella pneumoniae/drug effects , Lung/metabolism , Pulmonary Surfactants/pharmacology , Anti-Bacterial Agents/metabolism , Bronchoalveolar Lavage Fluid/chemistry , Drug Synergism , Humans , Immunity, Innate/physiology , Klebsiella Infections/pathology , Klebsiella Infections/prevention & control , Klebsiella pneumoniae/immunology , Lung/chemistry , Lung/immunology , Lung/microbiology , Microbial Sensitivity Tests , Pulmonary Surfactant-Associated Protein A/isolation & purification , Pulmonary Surfactant-Associated Protein A/metabolism , Pulmonary Surfactant-Associated Protein A/pharmacology , Pulmonary Surfactants/isolation & purification , Pulmonary Surfactants/metabolism , Respiratory Tract Infections/pathology , Respiratory Tract Infections/prevention & control
3.
Am J Respir Cell Mol Biol ; 62(4): 466-478, 2020 04.
Article in English | MEDLINE | ID: mdl-31922895

ABSTRACT

Surfactant protein (SP)-C deficiency is found in samples from patients with idiopathic pulmonary fibrosis, especially in familial forms of this disease. We hypothesized that SP-C may contribute to fibrotic remodeling in aging mice and alveolar lipid homeostasis. For this purpose, we analyzed lung function, alveolar dynamics, lung structure, collagen content, and expression of genes related to lipid and cholesterol metabolism of aging SP-C knockout mice. In addition, in vitro experiments with an alveolar macrophage cell line exposed to lipid vesicles with or without cholesterol and/or SP-C were performed. Alveolar dynamics showed progressive alveolar derecruitment with age and impaired oxygen saturation. Lung structure revealed that decreasing volume density of alveolar spaces was accompanied by increasing of the ductal counterparts. Simultaneously, septal wall thickness steadily increased, and fibrotic wounds appeared in lungs from the age of 50 weeks. This remarkable phenotype is unique to the 129Sv strain, which has an increased absorption of cholesterol, linking the accumulation of cholesterol and the absence of SP-C to a fibrotic remodeling process. The findings of this study suggest that overall loss of SP-C results in an age-dependent, complex, heterogeneous phenotype characterized by a combination of overdistended air spaces and fibrotic wounds that resembles combined emphysema and pulmonary fibrosis in patients with idiopathic pulmonary fibrosis. Addition of SP-C to cholesterol-laden lipid vesicles enhanced the expression of cholesterol metabolism and transport genes in an alveolar macrophage cell line, identifying a potential new lipid-protein axis involved in lung remodeling.


Subject(s)
Airway Remodeling/physiology , Cholesterol/metabolism , Idiopathic Pulmonary Fibrosis/metabolism , Protein C/metabolism , Pulmonary Surfactants/metabolism , Aged , Animals , Emphysema/metabolism , Humans , Lipid Metabolism/physiology , Lung/metabolism , Macrophages, Alveolar/metabolism , Male , Mice , Mice, Knockout , Pulmonary Alveoli/metabolism
4.
Int J Mol Sci ; 20(17)2019 Aug 30.
Article in English | MEDLINE | ID: mdl-31480246

ABSTRACT

High surface tension at the alveolar air-liquid interface is a typical feature of acute and chronic lung injury. However, the manner in which high surface tension contributes to lung injury is not well understood. This study investigated the relationship between abnormal alveolar micromechanics, alveolar epithelial injury, intra-alveolar fluid properties and remodeling in the conditional surfactant protein B (SP-B) knockout mouse model. Measurements of pulmonary mechanics, broncho-alveolar lavage fluid (BAL), and design-based stereology were performed as a function of time of SP-B deficiency. After one day of SP-B deficiency the volume of alveolar fluid V(alvfluid,par) as well as BAL protein and albumin levels were normal while the surface area of injured alveolar epithelium S(AEinjure,sep) was significantly increased. Alveoli and alveolar surface area could be recruited by increasing the air inflation pressure. Quasi-static pressure-volume loops were characterized by an increased hysteresis while the inspiratory capacity was reduced. After 3 days, an increase in V(alvfluid,par) as well as BAL protein and albumin levels were linked with a failure of both alveolar recruitment and airway pressure-dependent redistribution of alveolar fluid. Over time, V(alvfluid,par) increased exponentially with S(AEinjure,sep). In conclusion, high surface tension induces alveolar epithelial injury prior to edema formation. After passing a threshold, epithelial injury results in vascular leakage and exponential accumulation of alveolar fluid critically hampering alveolar recruitability.


Subject(s)
Alveolar Epithelial Cells/pathology , Bronchoalveolar Lavage Fluid/chemistry , Pulmonary Surfactant-Associated Protein B/deficiency , Acinar Cells/pathology , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/ultrastructure , Animals , Biomechanical Phenomena , Doxycycline/pharmacology , Female , Lung/drug effects , Lung/physiopathology , Lung/ultrastructure , Mice, Knockout , Models, Biological , Pulmonary Surfactant-Associated Protein B/metabolism , Structure-Activity Relationship , Surface Tension
6.
J Immunol ; 194(12): 5635-43, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25980009

ABSTRACT

Allergic asthma is a chronic inflammatory disorder that affects ∼20% of the population worldwide. Microarray analyses of nasal epithelial cells from acute asthmatic patients detected a 50% decrease in expression of Stard7, an intracellular phosphatidylcholine transport protein. To determine whether loss of Stard7 expression promotes allergic responses, mice were generated in which one allele of the Stard7 locus was globally disrupted (Stard7 (+/-) mice). OVA sensitization and challenge of Stard7(+/-) mice resulted in a significant increase in pulmonary inflammation, mucous cell metaplasia, airway hyperresponsiveness, and OVA-specific IgE compared with OVA-sensitized/challenged wild-type (WT) mice. This exacerbation was largely Th2-mediated with a significant increase in CD4(+)IL-13(+) T cells and IL-4, IL-5, and IL-13 cytokines. The loss of Stard7 was also associated with increased lung epithelial permeability and activation of proinflammatory dendritic cells in sensitized and/or challenged Stard7 (+/-) mice. Notably, OVA-pulsed dendritic cells from Stard7(+/-) mice were sufficient to confer an exaggerated allergic response in OVA-challenged WT mice, although airway hyperresponsiveness was greater in Stard7(+/-) recipients compared with WT recipients. Enhanced allergic responses in the lung were accompanied by age-dependent development of spontaneous atopic dermatitis. Overall, these data suggest that Stard7 is an important component of a novel protective pathway in tissues exposed to the extracellular environment.


Subject(s)
Carrier Proteins/genetics , Haploinsufficiency , Hypersensitivity/genetics , Hypersensitivity/immunology , Lung/immunology , Skin/immunology , Adoptive Transfer , Animals , Cytokines/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dermatitis/genetics , Dermatitis/immunology , Dermatitis/metabolism , Disease Models, Animal , Disease Progression , Female , Gene Deletion , Lung/metabolism , Lung/pathology , Male , Mice , Mice, Knockout , Models, Biological , Ovalbumin/adverse effects , Ovalbumin/immunology , Permeability , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Skin/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism
7.
J Immunol ; 195(4): 1628-36, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26163587

ABSTRACT

The anionic antimicrobial peptide SP-B(N), derived from the N-terminal saposin-like domain of the surfactant protein (SP)-B proprotein, and SP-A are lung anti-infective proteins. SP-A-deficient mice are more susceptible than wild-type mice to lung infections, and bacterial killing is enhanced in transgenic mice overexpressing SP-B(N). Despite their potential anti-infective action, in vitro studies indicate that several microorganisms are resistant to SP-A and SP-B(N). In this study, we test the hypothesis that these proteins act synergistically or cooperatively to strengthen each other's microbicidal activity. The results indicate that the proteins acted synergistically in vitro against SP-A- and SP-B(N)-resistant capsulated Klebsiella pneumoniae (serotype K2) at neutral pH. SP-A and SP-B(N) were able to interact in solution (Kd = 0.4 µM), which enabled their binding to bacteria with which SP-A or SP-B(N) alone could not interact. In vivo, we found that treatment of K. pneumoniae-infected mice with SP-A and SP-B(N) conferred more protection against K. pneumoniae infection than each protein individually. SP-A/SP-B(N)-treated infected mice showed significant reduction of bacterial burden, enhanced neutrophil recruitment, and ameliorated lung histopathology with respect to untreated infected mice. In addition, the concentrations of inflammatory mediators in lung homogenates increased early in infection in contrast with the weak inflammatory response of untreated K. pneumoniae-infected mice. Finally, we found that therapeutic treatment with SP-A and SP-B(N) 6 or 24 h after bacterial challenge conferred significant protection against K. pneumoniae infection. These studies show novel anti-infective pathways that could drive development of new strategies against pulmonary infections.


Subject(s)
Pulmonary Surfactant-Associated Proteins/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Antimicrobial Cationic Peptides/pharmacology , Cytokines/metabolism , Disease Models, Animal , Drug Synergism , Humans , Hydrogen-Ion Concentration , Klebsiella Infections/immunology , Klebsiella Infections/metabolism , Klebsiella Infections/microbiology , Klebsiella pneumoniae/drug effects , Klebsiella pneumoniae/immunology , Lung/drug effects , Lung/immunology , Lung/microbiology , Lung/pathology , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Knockout , Neutrophil Infiltration , Protein Binding , Pulmonary Surfactant-Associated Protein A/genetics , Pulmonary Surfactant-Associated Protein A/metabolism , Pulmonary Surfactant-Associated Protein A/pharmacology , Pulmonary Surfactant-Associated Protein B/genetics , Pulmonary Surfactant-Associated Protein B/metabolism , Pulmonary Surfactant-Associated Protein B/pharmacology , Pulmonary Surfactant-Associated Proteins/genetics , Pulmonary Surfactant-Associated Proteins/pharmacology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology
9.
Am J Respir Cell Mol Biol ; 53(1): 1-7, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25932959

ABSTRACT

Gas exchange after birth is entirely dependent on the remarkable architecture of the alveolus, its formation and function being mediated by the interactions of numerous cell types whose precise positions and activities are controlled by a diversity of signaling and transcriptional networks. In the later stages of gestation, alveolar epithelial cells lining the peripheral lung saccules produce increasing amounts of surfactant lipids and proteins that are secreted into the airspaces at birth. The lack of lung maturation and the associated lack of pulmonary surfactant in preterm infants causes respiratory distress syndrome, a common cause of morbidity and mortality associated with premature birth. At the time of birth, surfactant homeostasis begins to be established by balanced processes involved in surfactant production, storage, secretion, recycling, and catabolism. Insights from physiology and engineering made in the 20th century enabled survival of newborn infants requiring mechanical ventilation for the first time. Thereafter, advances in biochemistry, biophysics, and molecular biology led to an understanding of the pulmonary surfactant system that made possible exogenous surfactant replacement for the treatment of preterm infants. Identification of surfactant proteins, cloning of the genes encoding them, and elucidation of their roles in the regulation of surfactant synthesis, structure, and function have provided increasing understanding of alveolar homeostasis in health and disease. This Perspective seeks to consider developmental aspects of the pulmonary surfactant system and its importance in the pathogenesis of acute and chronic lung diseases related to alveolar homeostasis.


Subject(s)
Homeostasis , Pulmonary Alveoli , Pulmonary Surfactants/metabolism , Humans , Infant, Newborn , Infant, Premature , Pulmonary Alveoli/embryology , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/pathology , Respiration, Artificial , Respiratory Distress Syndrome, Newborn/embryology , Respiratory Distress Syndrome, Newborn/genetics , Respiratory Distress Syndrome, Newborn/metabolism , Respiratory Distress Syndrome, Newborn/pathology , Respiratory Distress Syndrome, Newborn/therapy
10.
J Lipid Res ; 56(7): 1370-9, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26022805

ABSTRACT

The surfactant proteins (SPs), SP-B and SP-C, are important components of pulmonary surfactant involved in the reduction of alveolar surface tension. Quantification of SP-B and SP-C in surfactant drugs is informative for their quality control and the evaluation of their biological activity. Western blot analysis enabled the quantification of SP-B, but not SP-C, in surfactant drugs. Here, we report a new procedure involving chemical treatments and LC-MS to analyze SP-C peptides. The procedure enabled qualitative analysis of SP-C from different species with discrimination of the palmitoylation status and the artificial modifications that occur during handling and/or storage. In addition, the method can be used to estimate the total amount of SP-C in pulmonary surfactant drugs. The strategy described here might serve as a prototype to establish analytical methods for peptides that are extremely hydrophobic and behave like lipids. The new method provides an easy measurement of SP-C from various biological samples, which will help the characterization of various experimental animal models and the quality control of surfactant drugs, as well as diagnostics of human samples.


Subject(s)
Chromatography, Liquid/methods , Lipoylation , Mass Spectrometry/methods , Pulmonary Surfactant-Associated Protein B/analysis , Pulmonary Surfactant-Associated Protein B/chemistry , Pulmonary Surfactant-Associated Protein C/analysis , Pulmonary Surfactant-Associated Protein C/chemistry , Animals , Blotting, Western , Cattle , Humans , Mice , Peptide Fragments/analysis , Surface-Active Agents/chemistry
11.
Am J Respir Crit Care Med ; 189(2): 214-22, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24160862

ABSTRACT

The median survival of patients with idiopathic pulmonary fibrosis (IPF) continues to be approximately 3 years from the time of diagnosis, underscoring the lack of effective medical therapies for this disease. In the United States alone, approximately 40,000 patients die of this disease annually. In November 2012, the NHLBI held a workshop aimed at coordinating research efforts and accelerating the development of IPF therapies. Basic, translational, and clinical researchers gathered with representatives from the NHLBI, patient advocacy groups, pharmaceutical companies, and the U.S. Food and Drug Administration to review the current state of IPF research and identify priority areas, opportunities for collaborations, and directions for future research. The workshop was organized into groups that were tasked with assessing and making recommendations to promote progress in one of the following six critical areas of research: (1) biology of alveolar epithelial injury and aberrant repair; (2) role of extracellular matrix; (3) preclinical modeling; (4) role of inflammation and immunity; (5) genetic, epigenetic, and environmental determinants; (6) translation of discoveries into diagnostics and therapeutics. The workshop recommendations provide a basis for directing future research and strategic planning by scientific, professional, and patient communities and the NHLBI.


Subject(s)
Idiopathic Pulmonary Fibrosis , Animals , Biomedical Research/trends , Disease Models, Animal , Extracellular Matrix/pathology , Genetic Predisposition to Disease , Humans , Idiopathic Pulmonary Fibrosis/diagnosis , Idiopathic Pulmonary Fibrosis/physiopathology , Idiopathic Pulmonary Fibrosis/therapy , Inflammation/immunology , Mice , Pulmonary Alveoli/pathology , Respiratory Mucosa/pathology
12.
Am J Respir Cell Mol Biol ; 51(3): 400-12, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24661119

ABSTRACT

Autophagy contributes to cellular homeostasis through metabolite recycling and degradation of cytotoxic protein aggregates and damaged organelles. Although recent studies have established that the requirement for basal autophagy is largely tissue specific, the importance of autophagy for alveolar epithelial cell homeostasis remains an important knowledge gap. In the present study we generated two mouse models, with > 90% or > 50% recombination at the Atg5 locus in the distal respiratory epithelium, to assess the effect of dose-dependent decreases in autophagy on alveolar homeostasis. A 90% decrease in autophagy was well tolerated in young adult mice but resulted in alveolar septal thickening and altered lung mechanics in aged animals, consistent with accumulation of damage over time. By comparison, a 50% decrease in autophagy had no effect on alveolar structure or function throughout the murine life span, indicating that basal autophagy in this compartment exceeds that required for homeostasis. A 50% decrease in autophagy in the bronchoalveolar epithelium significantly attenuated influenza A/H3N2 viral replication, leading to improved lung structure and function and reduced morbidity and mortality after infection. The reserve of autophagic capacity in the alveolar epithelium may provide a niche for replication of influenza A virus.


Subject(s)
Autophagy , Influenza A virus/physiology , Orthomyxoviridae Infections/virology , Pulmonary Alveoli/pathology , Pulmonary Alveoli/virology , Aging , Animals , Autophagy-Related Protein 5 , Body Weight , Female , Gene Deletion , Homeostasis , Lung/pathology , Male , Mice , Microtubule-Associated Proteins/genetics , Phenotype , Respiratory Mucosa/virology , Time Factors , Virus Replication
13.
Am J Respir Crit Care Med ; 186(10): 1014-24, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-23043085

ABSTRACT

RATIONALE: Hermansky-Pudlak syndrome (HPS) is a family of recessive disorders of intracellular trafficking defects that are associated with highly penetrant pulmonary fibrosis. Naturally occurring HPS mice reliably model important features of the human disease, including constitutive alveolar macrophage activation and susceptibility to profibrotic stimuli. OBJECTIVES: To decipher which cell lineage(s) in the alveolar compartment is the predominant driver of fibrotic susceptibility in HPS. METHODS: We used five different HPS and Chediak-Higashi mouse models to evaluate genotype-specific fibrotic susceptibility. To determine whether intrinsic defects in HPS alveolar macrophages cause fibrotic susceptibility, we generated bone marrow chimeras in HPS and wild-type mice. To directly test the contribution of the pulmonary epithelium, we developed a transgenic model with epithelial-specific correction of the HPS2 defect in an HPS mouse model. MEASUREMENTS AND MAIN RESULTS: Bone marrow transplantation experiments demonstrated that both constitutive alveolar macrophage activation and increased susceptibility to bleomycin-induced fibrosis were conferred by the genotype of the lung epithelium, rather than that of the bone marrow-derived, cellular compartment. Furthermore, transgenic epithelial-specific correction of the HPS defect significantly attenuated bleomycin-induced alveolar epithelial apoptosis, fibrotic susceptibility, and macrophage activation. Type II cell apoptosis was genotype specific, caspase dependent, and correlated with the degree of fibrotic susceptibility. CONCLUSIONS: We conclude that pulmonary fibrosis in naturally occurring HPS mice is driven by intracellular trafficking defects that lower the threshold for pulmonary epithelial apoptosis. Our findings demonstrate a pivotal role for the alveolar epithelium in the maintenance of alveolar homeostasis and regulation of alveolar macrophage activation.


Subject(s)
Genetic Predisposition to Disease , Hermanski-Pudlak Syndrome/genetics , Pulmonary Alveoli/physiopathology , Pulmonary Fibrosis/physiopathology , Respiratory Mucosa/physiopathology , Adaptor Protein Complex 3/genetics , Animals , Bleomycin/pharmacology , Hermanski-Pudlak Syndrome/complications , Hermanski-Pudlak Syndrome/physiopathology , Humans , Lung/pathology , Macrophage Activation , Macrophages, Alveolar/physiology , Mice , Mice, Knockout , Mutation , Protein Transport , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/complications , Pulmonary Fibrosis/pathology
14.
Am J Respir Cell Mol Biol ; 47(3): 324-31, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22461427

ABSTRACT

Mutations in the SFTPC gene, encoding surfactant protein-C (SP-C), are associated with interstitial lung disease (ILD). Knowledge of the intracellular fate of mutant SP-C is essential in the design of therapies to correct trafficking/processing of the proprotein, and to prevent the formation of cytotoxic aggregates. We assessed the potential of a chemical chaperone to correct the trafficking and processing of three disease-associated mutant SP-C proteins. HEK293 cells were stably transfected with wild-type (SP-C(WT)) or mutant (SP-C(L188Q), SP-C(Δexon4), or SP-C(I73T)) SP-C, and cell lines with a similar expression of SP-C mRNA were identified. The effects of the chemical chaperone 4-phenylbutyric acid (PBA) and lysosomotropic drugs on intracellular trafficking to the endolysosomal pathway and the subsequent conversion of SP-C proprotein to mature peptide were assessed. Despite comparable SP-C mRNA expression, proprotein concentrations varied greatly: SP-C(I73T) was more abundant than SP-C(WT) and was localized to the cell surface, whereas SP-C(Δexon4) was barely detectable. In contrast, SP-C(L188Q) and SP-C(WT) proprotein concentrations were comparable, and a small amount of SP-C(L188Q) was localized to the endolysosomal pathway. PBA treatment restored the trafficking and processing of SP-C(L188Q) to SP-C(WT) concentrations, but did not correct the mistrafficking of SP-C(I73T) or rescue SP-C(Δexon4). PBA treatment also promoted the aggregation of SP-C proproteins, including SP-C(L188Q). This study provides proof of the principle that a chemical chaperone can correct the mistrafficking and processing of a disease-associated mutant SP-C proprotein.


Subject(s)
Mutation , Phenylbutyrates/pharmacology , Pulmonary Surfactant-Associated Protein C/metabolism , Cell Line , Detergents/chemistry , Electrophoresis, Polyacrylamide Gel , Humans , Protein Processing, Post-Translational , Protein Transport , Pulmonary Surfactant-Associated Protein C/genetics , RNA, Messenger/genetics , Solubility
15.
Annu Rev Med ; 61: 105-19, 2010.
Article in English | MEDLINE | ID: mdl-19824815

ABSTRACT

The alveolar region of the lung creates an extensive epithelial surface that mediates the transfer of oxygen and carbon dioxide required for respiration after birth. Maintenance of pulmonary function depends on the function of type II epithelial cells that synthesize and secrete pulmonary surfactant lipids and proteins, reducing the collapsing forces created at the air-liquid interface in the alveoli. Genetic and acquired disorders associated with the surfactant system cause both acute and chronic lung disease. Mutations in the ABCA3, SFTPA, SFTPB, SFTPC, SCL34A2, and TERT genes disrupt type II cell function and/or surfactant homeostasis, causing neonatal respiratory failure and chronic interstitial lung disease. Defects in GM-CSF receptor function disrupt surfactant clearance, causing pulmonary alveolar proteinosis. Abnormalities in the surfactant system and disruption of type II cell homeostasis underlie the pathogenesis of pulmonary disorders previously considered idiopathic, providing the basis for improved diagnosis and therapies of these rare lung diseases.


Subject(s)
Lung Diseases/etiology , Pulmonary Surfactant-Associated Proteins/physiology , Adult , Child , Epithelial Cells/physiology , Humans , Infant , Lung Diseases/diagnosis , Lung Diseases/therapy , Macrophages, Alveolar/physiology , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/pathology , Pulmonary Alveoli/physiopathology
16.
J Immunol ; 184(2): 975-83, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-20007532

ABSTRACT

Surfactant protein B (SP-B) proprotein contains three saposin-like protein (SAPLIP) domains: a SAPLIP domain corresponding to the mature SP-B peptide is essential for lung function and postnatal survival; the function of SAPLIP domains in the N-terminal (SP-BN) and C-terminal regions of the proprotein is not known. In the current study, SP-BN was detected in the supernatant of mouse bronchoalveolar lavage fluid (BALF) and in nonciliated bronchiolar cells, alveolar type II epithelial cells, and alveolar macrophages. rSP-BN indirectly promoted the uptake of bacteria by macrophage cell lines and directly killed bacteria at acidic pH, consistent with a lysosomal, antimicrobial function. Native SP-BN isolated from BALF also killed bacteria but only at acidic pH; the bactericidal activity of BALF at acidic pH was completely blocked by SP-BN Ab. Transgenic mice overexpressing SP-BN and mature SP-B peptide had significantly decreased bacterial burden and increased survival following intranasal inoculation with bacteria. These findings support the hypothesis that SP-BN contributes to innate host defense of the lung by supplementing the nonoxidant antimicrobial defenses of alveolar macrophages.


Subject(s)
Anti-Bacterial Agents/pharmacology , Protein Precursors/immunology , Proteolipids/immunology , Animals , Anti-Bacterial Agents/isolation & purification , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/immunology , Hydrogen-Ion Concentration , Immunity, Innate , Klebsiella pneumoniae/immunology , Macrophages, Alveolar/microbiology , Mice , Mice, Transgenic , Protein Precursors/analysis , Protein Precursors/pharmacology , Protein Structure, Tertiary , Proteolipids/analysis , Proteolipids/pharmacology , Saposins , Staphylococcus aureus/immunology , Tissue Distribution
17.
Am J Perinatol ; 29(6): 419-28, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22399216

ABSTRACT

Lamellar body count (LBC) in amniotic fluid is a well-established method for assessing fetal lung maturity. However, the biogenesis and function of lamellar bodies (LBs) secreted into the amniotic fluid have not been formally assessed. We purified LBs from amniotic fluids obtained from term gestation pregnancies that had been determined to have mature LBC. Using tandem mass spectrometry, we identified 122 unique proteins in the LB preparations from the amniotic fluids. There was minimal overlap between the proteins identified in amniotic fluid LB and those reported for human epidermis LB. In contrast, there was >40% concordance with the proteome of rat lung LBs despite species differences. Classification of the identified proteins into functional bins demonstrated that the preponderance of amniotic fluid LB proteins was associated with host defense or anti-inflammatory functions. These data suggest that amniotic fluid LBs are derived from lung secretions and may play an important role in innate host defense of the fetus.


Subject(s)
Amniotic Fluid/chemistry , Epidermis/chemistry , Fetal Organ Maturity , Lung/chemistry , Proteome/analysis , Pulmonary Surfactants/analysis , Animals , Blotting, Western , Electrophoresis, Polyacrylamide Gel , Female , Humans , Infant, Newborn , Mass Spectrometry , Proteome/classification , Rats
18.
Physiology (Bethesda) ; 25(3): 132-41, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20551227

ABSTRACT

Pulmonary surfactant is an essential lipid-protein complex that stabilizes the respiratory units (alveoli) involved in gas exchange. Quantitative or qualitative derangements in surfactant are associated with severe respiratory pathologies. The integrated regulation of surfactant synthesis, secretion, and metabolism is critical for air breathing and, ultimately, survival. The goal of this review is to summarize our current understanding and highlight important knowledge gaps in surfactant homeostatic mechanisms.


Subject(s)
Lung Diseases/physiopathology , Models, Biological , Pulmonary Surfactants , Homeostasis/physiology , Humans , Pulmonary Gas Exchange/physiology
19.
J Cell Biol ; 172(3): 395-407, 2006 Jan 30.
Article in English | MEDLINE | ID: mdl-16449190

ABSTRACT

Mutations in the gene encoding SP-C (surfactant protein C; SFTPC) have been linked to interstitial lung disease (ILD) in children and adults. Expression of the index mutation, SP-C(Deltaexon4), in transiently transfected cells and type II cells of transgenic mice resulted in misfolding of the proprotein, activation of endoplasmic reticulum (ER) stress pathways, and cytotoxicity. In this study, we show that stably transfected cells adapted to chronic ER stress imposed by the constitutive expression of SP-C(Deltaexon4) via an NF-kappaB-dependent pathway. However, the infection of cells expressing SP-C(Deltaexon4) with respiratory syncytial virus resulted in significantly enhanced cytotoxicity associated with accumulation of the mutant proprotein, pronounced activation of the unfolded protein response, and cell death. Adaptation to chronic ER stress imposed by misfolded SP-C was associated with increased susceptibility to viral-induced cell death. The wide variability in the age of onset of ILD in patients with SFTPC mutations may be related to environmental insults that ultimately overwhelm the homeostatic cytoprotective response.


Subject(s)
Genetic Predisposition to Disease/genetics , Lung Diseases, Interstitial/genetics , Peptides/genetics , Protein Folding , Respiratory Syncytial Viruses , Animals , Apoptosis/drug effects , Caspase 3 , Caspases/metabolism , Cell Line , Cell Survival/drug effects , Cell Survival/genetics , DNA-Binding Proteins/metabolism , Down-Regulation/genetics , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/virology , Gene Expression/genetics , Gene Expression Profiling , Humans , I-kappa B Proteins/metabolism , Immunohistochemistry , Leupeptins/pharmacology , Lung Diseases, Interstitial/virology , Lysosomal Membrane Proteins/analysis , Mice , Mice, Transgenic , Mutation/genetics , NF-KappaB Inhibitor alpha , NF-kappa B p50 Subunit/antagonists & inhibitors , NF-kappa B p50 Subunit/metabolism , Nuclear Proteins/metabolism , Oligopeptides/analysis , Peptides/chemistry , Peptides/metabolism , Peptides/pharmacology , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors , Protein Sorting Signals , Pulmonary Surfactant-Associated Protein C , Regulatory Factor X Transcription Factors , Transcription Factors , Transfection , Up-Regulation/genetics
20.
JCI Insight ; 6(14)2021 07 22.
Article in English | MEDLINE | ID: mdl-34138759

ABSTRACT

Mutations in the gene SFTPC, encoding surfactant protein C (SP-C), are associated with interstitial lung disease in children and adults. To assess the natural history of disease, we knocked in a familial, disease-associated SFTPC mutation, L188Q (L184Q [LQ] in mice), into the mouse Sftpc locus. Translation of the mutant proprotein, proSP-CLQ, exceeded that of proSP-CWT in neonatal alveolar type 2 epithelial cells (AT2 cells) and was associated with transient activation of oxidative stress and apoptosis, leading to impaired expansion of AT2 cells during postnatal alveolarization. Differentiation of AT2 to AT1 cells was also inhibited in ex vivo organoid culture of AT2 cells isolated from LQ mice; importantly, treatment with antioxidant promoted alveolar differentiation. Upon completion of alveolarization, SftpcLQ expression was downregulated, leading to resolution of chronic stress responses; however, the failure to restore AT2 cell numbers resulted in a permanent loss of AT2 cells that was linked to decreased regenerative capacity in the adult lung. Collectively, these data support the hypothesis that susceptibility to disease in adult LQ mice is established during postnatal lung development, and they provide a potential explanation for the delayed onset of disease in patients with familial pulmonary fibrosis.


Subject(s)
Alveolar Epithelial Cells/pathology , Genetic Predisposition to Disease , Lung Diseases, Interstitial/genetics , Pulmonary Surfactant-Associated Protein C/genetics , Animals , Animals, Newborn , Cell Differentiation/genetics , Female , Gene Knock-In Techniques , Humans , Lung Diseases, Interstitial/pathology , Mice , Mutation
SELECTION OF CITATIONS
SEARCH DETAIL