Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 234
Filter
1.
Semin Fetal Neonatal Med ; 28(6): 101500, 2023 12.
Article in English | MEDLINE | ID: mdl-38036307

ABSTRACT

Genetic disorders of surfactant dysfunction are a rare cause of chronic, progressive or refractory respiratory failure in term and preterm infants. This review explores genetic mechanisms underpinning surfactant dysfunction, highlighting specific surfactant-associated genes including SFTPB, SFTPC, ABCA3, and NKX2.1. Pathogenic variants in these genes contribute to a range of clinical presentations and courses, from neonatal hypoxemic respiratory failure to childhood interstitial lung disease and even adult-onset pulmonary fibrosis. This review emphasizes the importance of early recognition, thorough phenotype assessment, and assessment of variant functionality as essential prerequisites for treatments including lung transplantation. We explore emerging treatment options, including personalized pharmacological approaches and gene therapy strategies. In conclusion, this comprehensive review offers valuable insights into the pathogenic mechanisms of genetic disorders of surfactant dysfunction, genetic fundamentals, available and emerging therapeutic options, and underscores the need for further research to develop personalized therapies for affected infants and children.


Subject(s)
Lung Diseases, Interstitial , Respiratory Insufficiency , Infant , Child , Adult , Infant, Newborn , Humans , Pulmonary Surfactant-Associated Protein B/genetics , Infant, Premature , Mutation , Lung Diseases, Interstitial/genetics , Lung Diseases, Interstitial/therapy
2.
Eur J Hum Genet ; 31(9): 1083-1087, 2023 09.
Article in English | MEDLINE | ID: mdl-37380697

ABSTRACT

Biallelic pathogenic variants in the surfactant protein (SP)-B gene (SFTPB) have been associated with fatal forms of interstitial lung diseases (ILD) in newborns and exceptional survival in young children. We herein report the cases of two related adults with pulmonary fibrosis due to a new homozygous SFTPB pathogenic variant, c.582G>A p.(Gln194=). In vitro transcript studies showed that this SFTPB synonymous pathogenic variant induces aberrant splicing leading to three abnormal transcripts with the preservation of the expression of a small proportion of normal SFTPB transcripts. Immunostainings on lung biopsies of the proband showed an almost complete loss of SP-B expression. This hypomorphic splice variant has thus probably allowed the patients' survival to adulthood while inducing an epithelial cell dysfunction leading to ILD. Altogether, this report shows that SFTPB pathogenic variants should be considered in atypical presentations and/or early-onset forms of ILD particularly when a family history is identified.


Subject(s)
Lung Diseases, Interstitial , Pulmonary Fibrosis , Adult , Child , Child, Preschool , Humans , Infant, Newborn , Lung Diseases, Interstitial/diagnosis , Lung Diseases, Interstitial/genetics , Pulmonary Fibrosis/genetics , Pulmonary Surfactant-Associated Protein B/genetics
3.
Bioengineered ; 13(1): 834-843, 2022 01.
Article in English | MEDLINE | ID: mdl-34898355

ABSTRACT

The rise of bioinformatics based on computer medicine provides a new method to reveal the complex biological data. This experiment is to explore the impacts of lipopolysaccharide on fetal lung developmental maturity and expressions of lung surfactant protein B (SP-B) and lung surfactant protein C (SP-C) in rats with gestational diabetes mellitus (GDM), thereby discussing the mechanism of developmental disorders in rats. Forty-eight conceived female rats were experimental subjects. Twenty-eight rats were randomly selected to construct the GDM models. All conceived rats underwent section on the 21st day of pregnancy. The ultrastructure of alveolar type II epithelial cells and the morphology of lung tissue were observed under a microscope. The protein localization and expression of SP-B and SP-C were determined by immunohistochemistry; the protein levels of SP-B and SP-C were determined by Western blot. Blood glucose and body weight of the GDM group were higher than those of the control group; the number of alveoli and alveolar area in the GDM group was lower than those in the control group; the alveolar interval in the GDM group was significantly higher than that in the control group (P < 0.05). The average absorbance of SP-B and SP-C in fetal lung tissue was significantly lower in the GDM group than that in the control group (P < 0.01). Changes in fetal lung tissue structure of rats were related to SP-B and SP-C, which was one of the main factors that affected the maturation of fetal lung tissue.


Subject(s)
Diabetes, Gestational/metabolism , Lipopolysaccharides/adverse effects , Lung/embryology , Lung/pathology , Peptides/metabolism , Pulmonary Surfactant-Associated Protein B/metabolism , Animals , Body Weight , Case-Control Studies , Diabetes, Gestational/blood , Diabetes, Gestational/genetics , Disease Models, Animal , Female , Gene Expression Regulation, Developmental/drug effects , Lung/drug effects , Lung/ultrastructure , Male , Peptides/genetics , Pregnancy , Pulmonary Surfactant-Associated Protein B/genetics , Random Allocation , Rats
4.
Protein Pept Lett ; 28(9): 1033-1042, 2021.
Article in English | MEDLINE | ID: mdl-33645472

ABSTRACT

BACKGROUND: Pulmonary surfactant dysfunction is an important pathological factor in acute respiratory distress syndrome (ARDS) and pulmonary fibrosis (PF). OBJECTIVE: In this study, the characteristics of recombinant mature surfactant protein B (SP-B) and reteplase (rPA) fusion protein maintaining good pulmonary surface activity and rPA fibrinolytic activity in acute lung injury cell model were studied. METHODS: We studied the characteristics of SP-B fusion expression, cloned rPA gene and N-terminal rPA/C-terminal SP-B co-expression gene, and constructed them into eukaryotic expression vector pEZ-M03 to obtain recombinant plasmids pEZ-rPA and pEZ-rPA/SP-B. The recombinant plasmids was transfected into Chinese hamster ovary (CHO) K1 cells and the expression products were analyzed by Western Blot. Lipopolysaccharide (LPS) was used to induce CCL149 (an alveolar epithelial cell line) cell injury model. Fluorescence staining of rPA and rPA/SP-B was carried out with the enhanced green fluorescent protein (eGFP) that comes with pEZ-M03; the cell Raman spectroscopy technique was used to analyze the interaction between rPA/SP-B fusion protein and the phospholipid structure of cell membrane in CCL149 cells. The enzyme activity of rPA in the fusion protein was determined by fibrin-agarose plate method. RESULTS: The rPA/SP-B fusion protein was successfully expressed. In the CCL149 cell model of acute lung injury (ALI), the green fluorescence of rPA/SP-B is mainly distributed on the CCL149 cell membrane. The rPA/SP-B fusion protein can reduce the disorder of phospholipid molecules and reduce cell membrane damage. The enzyme activity of rPA/SP-B fusion protein was 3.42, and the fusion protein still had good enzyme activity. CONCLUSION: The recombinant eukaryotic plasmid pEZ-rPA/SP-B is constructed and can be expressed in the eukaryotic system. Studies have shown that rPA/SP-B fusion protein maintains good SP-B lung surface activity and rPA enzyme activity in acute lung injury cell model.


Subject(s)
Epithelial Cells/metabolism , Pulmonary Alveoli/metabolism , Pulmonary Surfactant-Associated Protein B , Recombinant Fusion Proteins , Respiratory Distress Syndrome/drug therapy , Tissue Plasminogen Activator , Animals , CHO Cells , Cricetulus , Humans , Lipopolysaccharides/toxicity , Pulmonary Surfactant-Associated Protein B/biosynthesis , Pulmonary Surfactant-Associated Protein B/chemistry , Pulmonary Surfactant-Associated Protein B/genetics , Pulmonary Surfactant-Associated Protein B/pharmacology , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Respiratory Distress Syndrome/chemically induced , Respiratory Distress Syndrome/metabolism , Tissue Plasminogen Activator/biosynthesis , Tissue Plasminogen Activator/chemistry , Tissue Plasminogen Activator/genetics , Tissue Plasminogen Activator/pharmacology
5.
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
6.
Nat Commun ; 11(1): 3929, 2020 08 06.
Article in English | MEDLINE | ID: mdl-32764559

ABSTRACT

Surfactant protein B (SP-B) deficiency is an autosomal recessive disorder that impairs surfactant homeostasis and manifests as lethal respiratory distress. A compelling argument exists for gene therapy to treat this disease, as de novo protein synthesis of SP-B in alveolar type 2 epithelial cells is required for proper surfactant production. Here we report a rationally designed adeno-associated virus (AAV) 6 capsid that demonstrates efficiency in lung epithelial cell transduction based on imaging and flow cytometry analysis. Intratracheal administration of this vector delivering murine or human proSFTPB cDNA into SP-B deficient mice restores surfactant homeostasis, prevents lung injury, and improves lung physiology. Untreated SP-B deficient mice develop fatal respiratory distress within two days. Gene therapy results in an improvement in median survival to greater than 200 days. This vector also transduces human lung tissue, demonstrating its potential for clinical translation against this lethal disease.


Subject(s)
Genetic Therapy/methods , Genetic Vectors , Parvovirinae/genetics , Pulmonary Alveolar Proteinosis/congenital , Pulmonary Surfactant-Associated Protein B/deficiency , Animals , Animals, Newborn , Cell Line , Dependovirus , Disease Models, Animal , Female , Gene Expression , HEK293 Cells , Humans , Lung/metabolism , Lung/pathology , Male , Mice , Mice, Transgenic , Protein Precursors/genetics , Proteolipids/genetics , Pulmonary Alveolar Proteinosis/genetics , Pulmonary Alveolar Proteinosis/metabolism , Pulmonary Alveolar Proteinosis/therapy , Pulmonary Surfactant-Associated Protein B/genetics , Pulmonary Surfactant-Associated Protein B/metabolism , Pulmonary Surfactant-Associated Proteins/genetics , Transduction, Genetic
7.
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
8.
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
9.
Shock ; 54(4): 507-519, 2020 10.
Article in English | MEDLINE | ID: mdl-31851120

ABSTRACT

Surfactant protein B (SP-B) is essential for life and plays critical roles in host defense and lowering alveolar surface tension. A single-nucleotide polymorphism (SNP rs1130866) of human SP-B (hSP-B) alters the N-linked glycosylation, thus presumably affecting SP-B function. This study has investigated the regulatory roles of hSP-B genetic variants on lung injury in pneumonia-induced sepsis. METHODS: Wild-type (WT) FVB/NJ and humanized transgenic SP-B-T and SP-B-C mice (expressing either hSP-B C or T allele without mouse SP-B gene) were infected intratracheally with 50 µL (4 × 10 colony-forming units [CFUs]/mouse) Pseudomonas aeruginosa Xen5 or saline, and then killed 24 or 48 h after infection. Bacterial dynamic growths were monitored from 0 to 48 h postinfection by in vivo imaging. Histopathological, cellular, and molecular changes of lung tissues and bronchoalveolar lavage fluid (BALF) were analyzed. Surface tension of surfactants was determined with constrained drop surfactometry. RESULTS: SP-B-C mice showed higher bioluminescence and CFUs, increased inflammation and mortality, the higher score of lung injury, and reduced numbers of lamellar bodies in type II cells compared with SP-B-T or WT (P < 0.05). Minimum surface tension increased dramatically in infected mice (P < 0.01) with the order of SP-B-C > SP-B-T > WT. Levels of multiple cytokines in the lung of infected SP-B-C were higher than those of SP-B-T and WT (P < 0.01). Furthermore, compared with SP-B-T or WT, SP-B-C exhibited lower SP-B, higher NF-κB and NLRP3 inflammasome activation, and higher activated caspase-3. CONCLUSIONS: hSP-B variants differentially regulate susceptibility through modulating the surface activity of surfactant, cell death, and inflammatory signaling in sepsis.


Subject(s)
Pneumonia/metabolism , Pneumonia/microbiology , Pseudomonas aeruginosa/pathogenicity , Pulmonary Surfactant-Associated Protein B/genetics , Sepsis/metabolism , Sepsis/microbiology , Animals , Enzyme-Linked Immunosorbent Assay , Genetic Predisposition to Disease/genetics , Humans , In Situ Nick-End Labeling , Inflammation/metabolism , Inflammation/microbiology , Mice , Microscopy, Electron, Transmission
11.
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
12.
Sci Rep ; 9(1): 13450, 2019 09 17.
Article in English | MEDLINE | ID: mdl-31530844

ABSTRACT

Surfactant protein B (SFTPB) deficiency is a fatal disease affecting newborn infants. Surfactant is produced by alveolar type II cells which can be differentiated in vitro from patient specific induced pluripotent stem cell (iPSC)-derived lung organoids. Here we show the differentiation of patient specific iPSCs derived from a patient with SFTPB deficiency into lung organoids with mesenchymal and epithelial cell populations from both the proximal and distal portions of the human lung. We alter the deficiency by infecting the SFTPB deficient iPSCs with a lentivirus carrying the wild type SFTPB gene. After differentiating the mutant and corrected cells into lung organoids, we show expression of SFTPB mRNA during endodermal and organoid differentiation but the protein product only after organoid differentiation. We also show the presence of normal lamellar bodies and the secretion of surfactant into the cell culture medium in the organoids of lentiviral infected cells. These findings suggest that a lethal lung disease can be targeted and corrected in a human lung organoid model in vitro.


Subject(s)
Genetic Therapy/methods , Induced Pluripotent Stem Cells/cytology , Lung/cytology , Pulmonary Alveolar Proteinosis/congenital , Pulmonary Surfactant-Associated Protein B/deficiency , Cell Differentiation , Epithelial Cells/physiology , Fibroblasts/cytology , Genetic Markers , Green Fluorescent Proteins/genetics , Humans , Induced Pluripotent Stem Cells/transplantation , Lentivirus/genetics , Organoids , Pulmonary Alveolar Proteinosis/genetics , Pulmonary Alveolar Proteinosis/therapy , Pulmonary Alveoli/cytology , Pulmonary Surfactant-Associated Protein B/genetics
13.
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
14.
Curr Med Sci ; 38(5): 847-852, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30341519

ABSTRACT

This study investigated the expression of lung surfactant proteins (SP-B and SP-C), and regulatory factors [forkhead box A2 (FOXA2) and nitrolyogenic FOXA2 (N-FOXA2)] in the fetal lung of rats with gestational diabetes mellitus (GDM) in order to study the mechanism of pulmonary dysplasia. The rat GDM model was established by using streptozotocin intraperitoneally in the first stage of pregnancy. There were 10 rats in the GDM group, and 10 healthy rats in normal control group without any treatment. Fetal lungs of two groups were taken at day 21 of pregnancy. Blood glucose levels of maternal rats and fetal rats were measured by Roche blood glucose meter. The histological changes in the fetal lung were observed under the light microscope in both groups. The SP-B, SP-C and FOXA2 were determined in the fetal lung of two groups immunohistochemically. The expression levels of SP-B, SP-C, total FOXA2, FOXA2 in nucleus (n-FOXA2), N-FOXA2 proteins were detected by Western blotting, and the relative expression levels of SP-B, SP-C, FOXA2 mRNA in the fetal lung of two groups were detected by RTPCR. The results showed that blood glucose levels of maternal rats and fetal rats in GDM group were higher than those in control group. The light microscope revealed fetal lung development retardation in GDM group. The expression of SP-B and SP-C in GDM group was significantly reduced as compared with control group (P<0.05). As compared with control group, the n-FOXA2 expression was significantly decreased in the fetal lung tissue, and N-FOXA2 was significantly increased in control group (P<0.05), but there was no significant changes in the total FOXA2 (P>0.05). It was concluded that GDM can cause fetal lung development and maturation disorders, and FOXA2 in fetal lung tissue decreases while nitrocellulose FOXA2 increases.


Subject(s)
Diabetes, Gestational/genetics , Hepatocyte Nuclear Factor 3-beta/genetics , Peptides/genetics , Pulmonary Surfactant-Associated Protein B/genetics , Animals , Blood Glucose , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , Diabetes, Gestational/blood , Diabetes, Gestational/pathology , Disease Models, Animal , Female , Gene Expression Regulation, Developmental/genetics , Humans , Lung/growth & development , Lung/metabolism , Lung/pathology , Pregnancy , Pulmonary Surfactant-Associated Proteins/blood , Pulmonary Surfactant-Associated Proteins/genetics , Rats
15.
Pediatr Res ; 84(3): 419-425, 2018 09.
Article in English | MEDLINE | ID: mdl-29538355

ABSTRACT

BACKGROUND: Mutations in the NK2 homeobox 1 (NKX2-1) gene are associated with lung disease in infants and children. We hypothesize that disruption of normal surfactant gene expression with these mutations contributes to the respiratory phenotypes observed. METHODS: To assess transactivational activity, cotransfection of luciferase reporter vectors containing surfactant protein B or C (SFTPB or SFTPC) promoters with NKX2-1 plasmids was performed and luciferase activity was measured. To assess the binding of mutated proteins to target DNA, electrophoretic mobility shift assays (EMSA) were performed using nuclear protein labeled with oligonucleotide probes representing NKX2-1 consensus binding sequences followed by gel electrophoresis. The effect of overexpression of wild-type (WT) and mutant NKX2-1 on SFTPB and SFTPC was evaluated with quantitative real-time PCR. RESULTS: Decreased transactivation of the SFTPB promoter by both mutants and decreased transactivation of the SFTPC promoter by the L197P mutation was observed. EMSA demonstrated decreased DNA binding of both mutations to NKX2-1 consensus binding sequences. Transfection of A549 cells with NKX2-1 expression vectors demonstrated decreased stimulation of SFTPB and SFTPC expression by mutant proteins compared with that of WT. CONCLUSION: Disruption of transcriptional activation of surfactant protein genes by these DNA-binding domain mutations is a plausible biological mechanism for disruption of surfactant function and subsequent respiratory distress.


Subject(s)
Mutation , Promoter Regions, Genetic , Pulmonary Surfactant-Associated Protein B/genetics , Pulmonary Surfactant-Associated Protein C/genetics , Thyroid Nuclear Factor 1/genetics , A549 Cells , Adolescent , Cell Line, Tumor , Child, Preschool , Exons , Gene Expression Profiling , Gene Expression Regulation , Genes, Homeobox , Humans , Male , Mutagenesis, Site-Directed , Phenotype , Protein Binding , Retrospective Studies , Transcriptional Activation
16.
Mol Reprod Dev ; 85(3): 227-235, 2018 03.
Article in English | MEDLINE | ID: mdl-29388718

ABSTRACT

Neonatal respiratory distress is a major mortality factor in cloned animals, but the pathogenesis of this disease is rarely investigated. In this study, four neonatal cloned cattle, born after full-term gestation, exhibited symptoms of neonatal respiratory distress syndrome (NRDS), which included symptoms of hyaline membrane disease as well as disordered surfactant homeostasis in their collapsed lungs. No differences in DNA methylation or histone modifications correlated with the suppressed SPB and SPC transcription observed in the cloned cattle group (p > 0.05), whereas TTF-1 occupancy at SPB and SPC promoter regions in cloned cattle was significantly reduced to 24% and 20% that of normal lungs, respectively (SPB, p < 0.05; SPC, p < 0.01). Decreased TTF1 expression, dysregulation of SPB and SPC transcription by TTF-1, and disordered proteolytic processing of Surfactant protein B precursor together potentially contribute to the disruption of surfactant homeostasis and NRDS in bovine clones. Elucidation of the associated mechanisms should facilitate the development of novel preventive or therapeutic strategies to reduce the mortality rate of cloned animals and to improve the efficiency of SCNT technology.


Subject(s)
DNA Methylation , Promoter Regions, Genetic , Respiratory Distress Syndrome, Newborn/veterinary , Animals , Cattle , Cloning, Organism , Female , Histones/metabolism , Nuclear Transfer Techniques , Pulmonary Surfactant-Associated Protein B/genetics , Pulmonary Surfactant-Associated Protein B/metabolism , Respiratory Distress Syndrome, Newborn/genetics , Respiratory Distress Syndrome, Newborn/metabolism , Thyroid Nuclear Factor 1/genetics , Thyroid Nuclear Factor 1/metabolism
17.
Am J Obstet Gynecol ; 218(3): 349.e1-349.e10, 2018 03.
Article in English | MEDLINE | ID: mdl-29274832

ABSTRACT

BACKGROUND: The efficacy of antenatal steroids for fetal lung maturation in the periviable period is not fully understood. OBJECTIVE: We sought to determine the lung maturational effects of antenatal steroids and inflammation in early gestation sheep fetuses, similar to the periviable period in human beings. STUDY DESIGN: Date-mated ewes with singleton fetuses were randomly assigned to 1 of 4 treatment groups (n = 8/group): (1) maternal intramuscular injection of betamethasone; (2) intraamniotic lipopolysaccharide; (3) betamethasone + lipopolysaccharide; and (4) intraamniotic + intramuscular saline (controls). Fetuses were delivered surgically 48 hours later at 94 days' gestation (63% term gestation) for comprehensive evaluations of lung maturation, and lung and systemic inflammation. RESULTS: Relative to controls, first, betamethasone increased the fetal lung air space to mesenchymal area ratio by 47% but did not increase the messenger RNAs for the surfactant proteins-B and -C that are important for surfactant function or increase the expression of pro-surfactant protein-C in the alveolar type II cells. Second, betamethasone increased expression of 1 of the 4 genes in surfactant lipid synthetic pathways. Third, betamethasone increased genes involved in epithelium sodium channel transport, but not sodium-potassium adenosine triphosphatase or Aquaporin 5. Fourth, lipopolysaccharide increased proinflammatory genes in the lung but did not effectively recruit activated inflammatory cells. Last, betamethasone incompletely suppressed lipopolysaccharide-induced lung inflammation. In the liver, betamethasone when given alone increased the expression of serum amyloid A3 and C-reactive protein messenger RNAs. CONCLUSION: Compared the more mature 125-day gestation sheep, antenatal steroids do not induce pulmonary surfactants during the periviable period, indicating a different response.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Betamethasone/pharmacology , Gene Expression/drug effects , Lung/embryology , Premature Birth/drug therapy , RNA, Messenger/metabolism , Vascular Endothelial Growth Factor A/genetics , Alveolar Epithelial Cells , Animals , C-Reactive Protein/genetics , Cell Count , Chorioamnionitis/chemically induced , Chorioamnionitis/drug therapy , Chorioamnionitis/genetics , Cytokines/genetics , Female , Glutathione Peroxidase/genetics , Inflammation/chemically induced , Inflammation/drug therapy , Lipopolysaccharides , Lung/metabolism , Male , Pregnancy , Prenatal Care , Protein Isoforms , Pulmonary Surfactant-Associated Protein B/genetics , Pulmonary Surfactant-Associated Protein C/genetics , Random Allocation , Receptors, Glucocorticoid/genetics , Serum Amyloid A Protein/genetics , Sheep , Superoxide Dismutase/genetics , Glutathione Peroxidase GPX1
18.
J Perinatol ; 38(4): 337-344, 2018 04.
Article in English | MEDLINE | ID: mdl-29255193

ABSTRACT

OBJECTIVE: Genetic surfactant dysfunction causes respiratory failure in term and near-term newborn infants, but little is known of such condition in prematures. We evaluated genetic surfactant dysfunction in premature newborn infants with severe RDS. PATIENTS AND METHODS: A total of 68 preterm newborn infants with gestational age ≤32 weeks affected by unusually severe RDS were analysed for mutations in SFTPB, SFTPC and ABCA3. Therapies included oxygen supplementation, nasal CPAP, different modalities of ventilatory support, administration of exogenous surfactant, inhaled nitric oxide and steroids. Molecular analyses were performed on genomic DNA extracted from peripheral blood and Sanger sequencing of whole gene coding regions and intron junctions. In one case histology and electron microscopy on lung tissue was performed. RESULTS: Heterozygous previously described rare or novel variants in surfactant proteins genes ABCA3, SFTPB and SFTPC were identified in 24 newborn infants. In total, 11 infants died at age of 2 to 6 months. Ultrastructural analysis of lung tissue of one infant showed features suggesting ABCA3 dysfunction. DISCUSSION: Rare or novel genetic variants in genes encoding surfactant proteins were identified in a large proportion (35%) of premature newborn infants with particularly severe RDS. We speculate that interaction of developmental immaturity of surfactant production in association with abnormalities of surfactant metabolism of genetic origin may have a synergic worsening phenotypic effect.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Mutation , Pulmonary Surfactant-Associated Protein B/genetics , Pulmonary Surfactant-Associated Protein C/genetics , Respiratory Distress Syndrome, Newborn/genetics , Female , Gene Expression Regulation/genetics , Genetic Association Studies , Genetic Predisposition to Disease , Gestational Age , Heterozygote , Humans , Infant , Infant, Newborn , Infant, Premature , Italy , Lung/ultrastructure , Male , Retrospective Studies
19.
J Matern Fetal Neonatal Med ; 31(22): 2965-2970, 2018 Nov.
Article in English | MEDLINE | ID: mdl-28738720

ABSTRACT

BACKGROUND: Respiratory distress syndrome (RDS) is a severe pulmonary disease predominantly affects preterm newborns. Polymorphisms of surfactant-protein genes have been mostly evaluated as the candidate contributors in genetics of RDS. However the results are divers in different studies. We aimed at investigating the association of surfactant protein B (SPB) gene 9306 A/G polymorphism (rs7316) with RDS development. METHOD: Three hundred and eighty newborns with gestational age of less than 34 weeks were included in a multicenter case-control study. Respiratory distress (RD) was scored according to Downes' scoring system. Polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) method was used for genotyping. RESULT: One hundred and eighty-four neonates showed RDS and 196 did not. Gestational age (GA) was significantly lower in the RDS group compared with the controls. AA genotype and A allele were found more frequently in the RDS group than the controls (96.2% versus 63.8% and 98.1% versus 80.6%, respectively) (p =.0001). CONCLUSIONS: This is the first report of association of SFTPB rs7316 polymorphism with RDS development in Iranian newborns. The current study suggests that GA <28-weeks is the most important factor in predisposition to RDS. Genetic background in terms of SP-B gene might be involved in predisposition to RDS in premature neonates.


Subject(s)
Pulmonary Surfactant-Associated Protein B/genetics , Respiratory Distress Syndrome, Newborn/genetics , Case-Control Studies , Female , Genetic Predisposition to Disease , Humans , Infant, Newborn , Infant, Premature , Iran/epidemiology , Male , Polymorphism, Genetic , Respiratory Distress Syndrome, Newborn/mortality
20.
J Clin Invest ; 127(12): 4314-4325, 2017 12 01.
Article in English | MEDLINE | ID: mdl-29083321

ABSTRACT

Adaptation to respiration at birth depends upon the synthesis of pulmonary surfactant, a lipid-protein complex that reduces surface tension at the air-liquid interface in the alveoli and prevents lung collapse during the ventilatory cycle. Herein, we demonstrated that the gene encoding a subunit of the endoplasmic reticulum membrane complex, EMC3, also known as TMEM111 (Emc3/Tmem111), was required for murine pulmonary surfactant synthesis and lung function at birth. Conditional deletion of Emc3 in murine embryonic lung epithelial cells disrupted the synthesis and packaging of surfactant lipids and proteins, impaired the formation of lamellar bodies, and induced the unfolded protein response in alveolar type 2 (AT2) cells. EMC3 was essential for the processing and routing of surfactant proteins, SP-B and SP-C, and the biogenesis of the phospholipid transport protein ABCA3. Transcriptomic, lipidomic, and proteomic analyses demonstrated that EMC3 coordinates the assembly of lipids and proteins in AT2 cells that is necessary for surfactant synthesis and function at birth.


Subject(s)
Alveolar Epithelial Cells/metabolism , Lipid Metabolism/physiology , Membrane Proteins/metabolism , Peptides/metabolism , Pulmonary Surfactant-Associated Protein B/metabolism , Respiration , Alveolar Epithelial Cells/cytology , Animals , Gene Deletion , Intercellular Signaling Peptides and Proteins , Membrane Proteins/genetics , Mice , Mice, Transgenic , Organ Specificity , Peptides/genetics , Pulmonary Surfactant-Associated Protein B/genetics , Pulmonary Surfactant-Associated Protein C
SELECTION OF CITATIONS
SEARCH DETAIL
...