Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
4.
Pediatr Res ; 79(1-1): 27-33, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26375473

ABSTRACT

BACKGROUND: Heterozygous mutations of SFTPC, the gene-encoding surfactant protein C (SP-C), result in interstitial lung disease (ILD). However, characterization of mutations located in the mature domain of precursor SP-C (proSP-C) is limited. This study examined the molecular pathogenesis of such a mutation of ILD. METHODS: We employed sequencing of SFTPC and established A549 cells stably expressing several proSP-C mutants. Histopathology and transmission electron microscopy (TEM) of lung tissue from a pediatric patient with ILD were assessed. Effects of mutant proSP-C were evaluated by western blotting, immunofluorescence, and TEM. RESULTS: Sequencing of SFTPC revealed a novel heterozygous mutation, c.163C>T (L55F). In lung tissue, abnormal localization of proSP-C was observed by immunohistochemistry, and small and dense lamellar bodies (LBs) in type II alveolar epithelial cells (AECs) were detected by TEM. TEM of A549 cells stably expressing proSP-C(L55F) displayed abnormal cytoplasmic organelles. ProSP-C(L55F) exhibited a band pattern similar to that of proSP-C(WT) for processed intermediates. Immunofluorescence studies demonstrated that proSP-C(L55F) partially colocalized in CD63-positive cytoplasmic vesicles of A549 cells, which was in contrast to proSP-C(WT). CONCLUSION: We detected a novel c.163C>T mutation located in the mature domain of SFTPC associated with ILD that altered the subcellular localization of proSP-C in A549 cells.


Subject(s)
Alveolar Epithelial Cells/ultrastructure , Lung Diseases, Interstitial/genetics , Mutation, Missense , Point Mutation , Pulmonary Alveolar Proteinosis/genetics , Pulmonary Surfactant-Associated Protein C/deficiency , Alveolar Epithelial Cells/chemistry , Amino Acid Substitution , Cell Line , Cytoplasmic Granules/chemistry , Exons/genetics , Female , Heterozygote , Humans , Infant , Lung Diseases, Interstitial/diagnostic imaging , Lung Diseases, Interstitial/pathology , Lung Diseases, Interstitial/surgery , Lung Transplantation , Lysosomes/chemistry , Microscopy, Electron , Protein Precursors/analysis , Protein Processing, Post-Translational , Protein Structure, Tertiary , Pulmonary Alveolar Proteinosis/diagnostic imaging , Pulmonary Alveolar Proteinosis/pathology , Pulmonary Alveolar Proteinosis/surgery , Pulmonary Alveoli/pathology , Pulmonary Surfactant-Associated Protein C/analysis , Pulmonary Surfactant-Associated Protein C/genetics , Pulmonary Surfactant-Associated Protein C/metabolism , Radiography , Recombinant Proteins/analysis , Sequence Analysis, DNA , Subcellular Fractions/chemistry , Transfection
5.
Pediatr Res ; 79(1-1): 34-41, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26375475

ABSTRACT

BACKGROUND: Children's interstitial lung diseases (chILD) comprise a broad spectrum of diseases. Besides the genetically defined surfactant dysfunction disorders, most entities pathologically involve the alveolar surfactant region, possibly affecting the surfactant proteins SP-B and SP-C. Therefore, our objective was to determine the value of quantitation of SP-B and SP-C levels in bronchoalveolar lavage fluid (BALF) for the diagnosis of chILD. METHODS: Levels of SP-B and SP-C in BALF from 302 children with chILD and in controls were quantified using western blotting. In a subset, single-nucleotide polymorphisms (SNPs) in the SFTPC promoter were genotyped by direct sequencing. RESULTS: While a lack of dimeric SP-B was found only in the sole subject with hereditary SP-B deficiency, low or absent SP-C was observed not only in surfactant dysfunction disorders but also in patients with other diffuse parenchymal lung diseases pathogenetically related to the alveolar surfactant region. Genetic analysis of the SFTPC promoter showed association of a single SNP with SP-C level. CONCLUSION: SP-B levels may be used for screening for SP-B deficiency, while low SP-C levels may point out diseases caused by mutations in TTF1, SFTPC, ABCA3, and likely in other genes involved in surfactant metabolism that remain to be identified. We conclude that measurement of levels of SP-B and SP-C was useful for the differential diagnosis of chILD, and for the precise molecular diagnosis, sequencing of the genes is necessary.


Subject(s)
Bronchoalveolar Lavage Fluid/chemistry , Lung Diseases, Interstitial/diagnosis , Pulmonary Surfactant-Associated Protein B/analysis , Pulmonary Surfactant-Associated Protein C/analysis , ATP-Binding Cassette Transporters/genetics , Adolescent , Bronchitis/genetics , Case-Control Studies , Child , Child, Preschool , Comorbidity , DNA-Binding Proteins/genetics , Developmental Disabilities/genetics , Female , Genetic Heterogeneity , Genotype , Humans , Immunologic Deficiency Syndromes/genetics , Infant , Lung Diseases, Interstitial/genetics , Male , Polymorphism, Single Nucleotide , Promoter Regions, Genetic/genetics , Protein Precursors/genetics , Proteolipids/genetics , Pulmonary Alveolar Proteinosis/diagnosis , Pulmonary Alveolar Proteinosis/genetics , Pulmonary Surfactant-Associated Protein B/deficiency , Pulmonary Surfactant-Associated Protein B/genetics , Pulmonary Surfactant-Associated Protein C/chemistry , Pulmonary Surfactant-Associated Protein C/deficiency , Pulmonary Surfactant-Associated Protein C/genetics , Sequence Analysis, DNA , Transcription Factors , Young Adult
6.
Eur Respir J ; 46(1): 197-206, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25657025

ABSTRACT

Patients with interstitial lung disease due to surfactant protein C (SFTPC) mutations are rare and not well characterised. We report on all subjects collected over a 15-year period in the kids-lung register with interstitial lung disease and a proven SFTPC mutation. We analysed clinical courses, interventions and outcomes, as well as histopathological and radiological interrelations. 17 patients (seven male) were followed over a median of 3 years (range 0.3-19). All patients were heterozygous carriers of autosomal dominant SFTPC mutations. Three mutations (p.L101P, p.E191 K and p.E191*) have not been described before in the context of surfactant protein C deficiency. Patients with alterations in the BRICHOS domain of the protein (amino acids 94-197) presented earlier. At follow-up, one patient was healthy (2 years), six patients were "sick-better" (2.8 years, range 0.8-19), seven patients were "sick-same" (6.5 years, 1.3-15.8) and three patients were "sick-worse" (0.3 years, 0.3-16.9). Radiological findings changed from ground-glass to increasing signs of fibrosis and cyst formation with increasing age. Empiric treatments had variable effects, also in patients with the same genotype. Prospective studies with randomised interventions are urgently needed and can best be performed in the framework of international registers.


Subject(s)
Lung Diseases, Interstitial/genetics , Mutation , Pulmonary Surfactant-Associated Protein C/deficiency , Pulmonary Surfactant-Associated Protein C/genetics , Adolescent , Biopsy , Bronchoalveolar Lavage , Child , Child, Preschool , Female , Follow-Up Studies , Genes, Dominant , Genotype , Heterozygote , Humans , Infant , Infant, Newborn , Male , Prospective Studies , Pulmonary Surfactant-Associated Protein B/metabolism , Pulmonary Surfactant-Associated Protein C/metabolism , Retrospective Studies
7.
Curr Pediatr Rev ; 10(2): 162-7, 2014.
Article in English | MEDLINE | ID: mdl-25088270

ABSTRACT

Surfactant replacement therapy is now the standard of care for infants with respiratory distress syndrome. As the understanding of surfactant structure and function has evolved, surfactant-associated proteins are now understood to be essential components of pulmonary surfactant. Their structural and functional diversity detail the complexity of their contributions to normal pulmonary physiology, and deficiency states result in significant pathology. Engineering synthetic surfactant protein constructs has been a major research focus for replacement therapies. This review highlights what is known about surfactant proteins and how this knowledge is pivotal for future advancements in treating respiratory distress syndrome as well as other pulmonary diseases characterized by surfactant deficiency or inactivation.


Subject(s)
Biological Products/metabolism , Lung/metabolism , Pulmonary Surfactant-Associated Proteins/metabolism , Pulmonary Surfactants/metabolism , Respiratory Distress Syndrome, Newborn/metabolism , Biological Products/therapeutic use , Genetic Predisposition to Disease , Humans , Infant , Infant, Newborn , Infant, Premature , Lung/physiopathology , Pulmonary Surfactant-Associated Protein A/deficiency , Pulmonary Surfactant-Associated Protein B/deficiency , Pulmonary Surfactant-Associated Protein C/deficiency , Pulmonary Surfactant-Associated Protein D/deficiency , Pulmonary Surfactant-Associated Proteins/therapeutic use , Pulmonary Surfactants/therapeutic use , Respiratory Distress Syndrome, Newborn/genetics , Respiratory Distress Syndrome, Newborn/physiopathology
8.
Orphanet J Rare Dis ; 9: 36, 2014 Mar 19.
Article in English | MEDLINE | ID: mdl-24642012

ABSTRACT

BACKGROUND: Rare chronic diseases of childhood are often complex and associated with multiple health issues. Such conditions present significant demands on health services, but the degree of these demands is seldom reported. This study details the utilisation of hospital services and associated costs in a single case of surfactant protein C deficiency, an example of childhood interstitial lung disease. METHODS: Hospital records and case notes for a single patient were reviewed. Costs associated with inpatient services were extracted from a paediatric hospital database. Actual costs were compared to cost estimates based on both disease/procedure-related cost averages for inpatient hospital episodes and a recently implemented Australian hospital funding algorithm (activity-based funding). RESULTS: To age 8 years and 10 months the child was a hospital inpatient for 443 days over 32 admissions. A total of 298 days were spent in paediatric intensive care. Investigations included 58 chest x-rays, 9 bronchoscopies, 10 lung function tests and 11 sleep studies. Comprehensive disease management failed to prevent respiratory decline and a lung transplant was required. Costs of inpatient care at three tertiary hospitals totalled $966,531 (Australian dollars). Disease- and procedure-related cost averages underestimated costs of paediatric inpatient services for this patient by 68%. An activity-based funding algorithm that is currently being adopted in Australia estimated the cost of hospital health service provision with more accuracy. CONCLUSIONS: Health service usage and inpatient costs for this case of rare chronic childhood respiratory disease were substantial. This case study demonstrates that disease- and procedure-related cost averages are insufficient to estimate costs associated with rare chronic diseases that require complex management. This indicates that the health service use for similar episodes of hospital care is greater for children with rare diseases than other children. The impacts of rare chronic childhood diseases should be considered when planning resources for paediatric health services.


Subject(s)
Cost of Illness , Hospitalization/economics , Lung Diseases, Interstitial/etiology , Pulmonary Alveolar Proteinosis/complications , Pulmonary Surfactant-Associated Protein C/deficiency , Algorithms , Australia , Humans , Lung Diseases, Interstitial/complications , Lung Diseases, Interstitial/economics , Male , Patient Admission/statistics & numerical data , Pulmonary Alveolar Proteinosis/economics , Pulmonary Surfactant-Associated Protein C/economics
9.
BMJ Case Rep ; 20142014 Mar 19.
Article in English | MEDLINE | ID: mdl-24648475

ABSTRACT

The clinical course and treatment in the first 2.5 years of life of a term-born girl with a severe onset of respiratory symptoms in the neonatal period caused by a p.Cys121Phe/C121F mutation in the gene of surfactant protein C (SFTPC) is described. During the first 9 months of life, she was mechanically ventilated. With methylprednisolone pulse therapy and oral prednisolone, she could eventually gradually be weaned from mechanical ventilation. At the age of 2.5 years, she is in a good clinical condition without any respiratory support and has a normal nutritional status and neurodevelopment. This clinical course with neonatal onset of respiratory insufficiency is remarkable since most patients with SFTPC mutations present with milder respiratory symptoms in the first years of life.


Subject(s)
Lung/diagnostic imaging , Pulmonary Surfactant-Associated Protein C/deficiency , Respiration, Artificial , Respiratory Distress Syndrome, Newborn/therapy , Ventilator Weaning , Child, Preschool , Disease Progression , Female , Humans , Infant , Infant, Newborn , Respiratory Distress Syndrome, Newborn/diagnostic imaging , Tomography, X-Ray Computed
10.
Pediatr Pulmonol ; 49(3): E66-8, 2014 Mar.
Article in English | MEDLINE | ID: mdl-23775869

ABSTRACT

We present an infant who was born premature at 23 weeks gestation with bronchopulmonary dysplasia and a SFTPC gene mutation, p.R167Q, who had a complicated neonatal course requiring 4 months of mechanical ventilation. Over time, his clinical course has improved, and he only requires oxygen by nasal cannula and low dose hydroxychloroquine, suggesting that p.R167Q mutation contributed to his clinical course and may manifest with a variable disease pattern making long-term prognostication difficult in the immediate newborn period.


Subject(s)
Bronchopulmonary Dysplasia/physiopathology , Pulmonary Alveolar Proteinosis/genetics , Pulmonary Surfactant-Associated Protein C/genetics , Anti-Inflammatory Agents/therapeutic use , Bronchopulmonary Dysplasia/complications , Bronchopulmonary Dysplasia/therapy , Disease Progression , Humans , Infant, Extremely Premature , Infant, Newborn , Male , Mutation , Pulmonary Alveolar Proteinosis/complications , Pulmonary Alveolar Proteinosis/physiopathology , Pulmonary Surfactant-Associated Protein C/deficiency , Respiration, Artificial
11.
J Perinatol ; 33(6): 492-4, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23719253

ABSTRACT

SFTPC (surfactant protein C) mutations resulting in SP-C deficiency causing ongoing respiratory failure in the neonatal period represent a rare entity. We report a full-term female infant who developed respiratory distress and respiratory failure shortly after birth. From the first day of life the infant was mechanically ventilated. Application of exogenous surfactant or cortisone did not lead to any clinical improvement. Genetic analysis identified a novel SFTPC mutation as the cause of her lung disease. The patient was diagnosed as heterozygous for a p.Cys121Gly/C121G substitution encoded by exon 4, which could not be detected in both parents. Experimental therapy with hydroxychloroquine resulted in a significant clinical improvement within 2 weeks time. Mechanical ventilation was no longer needed, and the patient was discharged without additional oxygen demand. The patient remained well under therapy till the age of 6 months. After that time, the therapy was successfully discontinued.


Subject(s)
Amino Acid Substitution/genetics , DNA Mutational Analysis , Exons/genetics , Hydroxychloroquine/therapeutic use , Pulmonary Surfactant-Associated Protein C/genetics , Respiratory Distress Syndrome, Newborn/genetics , Respiratory Distress Syndrome, Newborn/therapy , Respiratory Insufficiency/genetics , Respiratory Insufficiency/therapy , Cesarean Section , Cysteine/genetics , Female , Follow-Up Studies , Genetic Carrier Screening , Glycine/genetics , Humans , Infant , Infant, Newborn , Pulmonary Surfactant-Associated Protein C/deficiency , Respiratory Distress Syndrome, Newborn/diagnosis , Respiratory Insufficiency/diagnosis
12.
Arch Bronconeumol ; 49(5): 213-5, 2013 May.
Article in English, Spanish | MEDLINE | ID: mdl-23137777

ABSTRACT

We present the case of two twin brothers with surfactant protein C deficiency who were treated with hydroxychloroquine for three years, with apparent success. The exact physiopathology of this disease is not known and there is no specific treatment for it. There is merely news from a few previous descriptions in the literature about the use of hydroxychloroquine for surfactant protein C deficiency with satisfactory results. Two years after the treatment was withdrawn, the twins were evaluated once again: they presented no new infections, growth and general state were normal and chest CT showed a notable additional reduction in the interstitial pneumopathy. These data seem to cast some doubt on the efficacy of hydroxychloroquine, and they suggest that the clinical improvement was simply the natural evolution of the disease.


Subject(s)
Diseases in Twins/drug therapy , Hydroxychloroquine/therapeutic use , Pulmonary Alveolar Proteinosis/drug therapy , Pulmonary Surfactant-Associated Protein C/deficiency , Disease Progression , Diseases in Twins/genetics , Dyspnea/etiology , Failure to Thrive/etiology , Humans , Infant, Newborn , Male , Pulmonary Alveolar Proteinosis/complications , Pulmonary Alveolar Proteinosis/diagnostic imaging , Pulmonary Alveolar Proteinosis/genetics , Respiratory Insufficiency/etiology , Tomography, X-Ray Computed , Twins, Monozygotic
13.
Cell Transplant ; 21(7): 1477-92, 2012.
Article in English | MEDLINE | ID: mdl-22507554

ABSTRACT

Since current treatments for both acute and chronic lung diseases are less than ideal, there has been recent interest in the use of cell-based therapies for inflammatory lung disease. Specifically, human amnion epithelial cells (hAECs) have been shown to reduce bleomycin-induced lung injury and prevent subsequent loss of respiratory function, primarily through modulation of the host immune response. The precise mechanisms of this effect remain unclear. We aimed to investigate the potential of hAECs to mitigate bleomycin-induced lung injury in surfactant protein C deficient (Sftpc(−/−)) mice, which are highly susceptible to pulmonary injury as a result of impairment of macrophage function. Primary hAECs were administered to wild-type (Sftpc(+/+)) and Sftpc(−/−) mice 24 h after exposure to bleomycin. Compared to Sftpc(+/+) mice receiving bleomycin alone, Sftpc(+/+) mice administered hAECs 24 h after bleomycin exposure had decreased expression of proinflammatory genes, decreased macrophage and neutrophil infiltration, fibrosis, collagen content, and α-smooth muscle actin as well as a significant improvement in lung function. Compared to Sftpc(−/−) mice given bleomycin alone, Sftpc(−/−) mice administered hAECs 24 h after bleomycin did not have a decrease in inflammatory gene expression or a reduction in macrophage pulmonary infiltration. Subsequently, Sftpc(−/−) mice did not show any decrease in pulmonary fibrosis or improvement of lung function after hAEC administration. The ability of hAECs to mitigate bleomycin-induced lung injury is abolished in Sftpc(−/−) mice, suggesting that hAECs require normal host macrophage function to exert their reparative effects.


Subject(s)
Amnion/cytology , Epithelial Cells/cytology , Macrophages/cytology , Pulmonary Fibrosis/therapy , Actins/metabolism , Animals , Antibiotics, Antineoplastic/toxicity , Bleomycin/toxicity , Collagen/metabolism , Disease Models, Animal , Epithelial Cells/transplantation , Humans , Lung Injury/chemically induced , Lung Injury/physiopathology , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Knockout , Neutrophil Infiltration/physiology , Pulmonary Fibrosis/physiopathology , Pulmonary Surfactant-Associated Protein C/deficiency , Pulmonary Surfactant-Associated Protein C/genetics , Pulmonary Surfactant-Associated Protein C/metabolism
14.
Clin Chest Med ; 33(1): 95-110, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22365249

ABSTRACT

The interstitial lung diseases (ILDs), or diffuse parenchymal lung diseases, are a heterogeneous collection of more than 100 different pulmonary disorders that affect the tissue and spaces surrounding the alveoli. Patients affected by ILD usually present with shortness of breath or cough; for many, there is evidence of pulmonary restriction, decreased diffusion capacity, and radiographic appearance of alveolar and/or reticulonodular infiltrates. This article reviews the inherited ILDs, with a focus on the diseases that may be seen by pulmonologists caring for adult patients. The authors conclude by briefly discussing the utility of genetic testing in this population.


Subject(s)
Birt-Hogg-Dube Syndrome/genetics , Genetic Diseases, Inborn/genetics , Lung Diseases, Interstitial/genetics , Mutation , Adult , Calcinosis/genetics , Female , Genetic Diseases, Inborn/complications , Genetic Diseases, X-Linked/genetics , Heterozygote , Humans , INDEL Mutation , Kidney Neoplasms/genetics , Lung Diseases/genetics , Lung Diseases, Interstitial/complications , Lung Diseases, Interstitial/diagnostic imaging , Lung Diseases, Interstitial/pathology , Lung Diseases, Interstitial/therapy , Pedigree , Pneumothorax/etiology , Proto-Oncogene Proteins/genetics , Pulmonary Alveolar Proteinosis/genetics , Pulmonary Fibrosis/genetics , Pulmonary Surfactant-Associated Protein B/deficiency , Pulmonary Surfactant-Associated Protein B/genetics , Pulmonary Surfactant-Associated Protein C/deficiency , Pulmonary Surfactant-Associated Protein C/genetics , RNA/genetics , Radiography , Recurrence , Sarcoidosis, Pulmonary/genetics , Siblings , Telomerase/genetics , Tumor Suppressor Proteins/genetics
15.
Clin Chest Med ; 33(1): 165-78, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22365253

ABSTRACT

Cigarette smoke, a toxic collection of thousands of chemicals generated from combustion of tobacco, is recognized as the primary causative agent of certain diffuse interstitial and bronchiolar lung diseases. Most patients afflicted with these disorders are cigarette smokers, and smoking cessation has been shown to be capable of inducing disease remission and should occupy a pivotal role in the management of all smokers with these diffuse lung diseases. The role of pharmacotherapy with corticosteroids or other immunomodulating agents is not well established but may be considered in patients with progressive forms of smoking-related interstitial lung diseases.


Subject(s)
Lung Diseases, Interstitial/diagnosis , Lung Diseases, Interstitial/etiology , Lung/diagnostic imaging , Lung/pathology , Smoking/adverse effects , Acute Disease , Bronchiolitis/diagnosis , Bronchiolitis/etiology , Chronic Disease , Diagnosis, Differential , Histiocytosis, Langerhans-Cell/diagnosis , Histiocytosis, Langerhans-Cell/etiology , Humans , Lung/physiopathology , Lung Diseases, Interstitial/classification , Lung Diseases, Interstitial/diagnostic imaging , Lung Diseases, Interstitial/pathology , Lung Diseases, Interstitial/physiopathology , Pulmonary Alveolar Proteinosis/diagnosis , Pulmonary Alveolar Proteinosis/etiology , Pulmonary Eosinophilia/diagnosis , Pulmonary Eosinophilia/etiology , Pulmonary Fibrosis/diagnosis , Pulmonary Fibrosis/etiology , Pulmonary Surfactant-Associated Protein C/deficiency , Tomography, X-Ray Computed
16.
Pneumonol Alergol Pol ; 78(3): 224-8, 2010.
Article in Polish | MEDLINE | ID: mdl-20461691

ABSTRACT

Etiology and pathogenesis of the interstitial lung disease in children result from a heterogeneous group of infectious, immunological and metabolic factors. In children an important role plays a surfactant protein B and C deficiency. SP-C deficiency is determined by it's defective synthesis or impaired production of ABCA3 transporter, as well as with abnormalities within different metabolic pathways. In the paper clinical manifestation, radiological findings, molecular background and prognosis in interstitial lung diseases associated with SP-B and SP-C defects have been discussed.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Lung Diseases, Interstitial/etiology , Lung Diseases, Interstitial/metabolism , Pulmonary Surfactant-Associated Protein B/deficiency , Pulmonary Surfactant-Associated Protein C/deficiency , ATP-Binding Cassette Transporters/genetics , Child , Humans , Prognosis , Pulmonary Surfactant-Associated Protein B/genetics , Pulmonary Surfactant-Associated Protein B/metabolism , Pulmonary Surfactant-Associated Protein C/genetics , Pulmonary Surfactant-Associated Protein C/metabolism , Risk Factors
17.
Pneumonol Alergol Pol ; 78(3): 244-7, 2010.
Article in Polish | MEDLINE | ID: mdl-20461694

ABSTRACT

The interstitial lung disease in children is a group of disorders heterogeneous in its etiology and pathogenesis. In differential diagnosis infectious, immunological and metabolic factors must be taken into consideration, as well as a genetically determined surfactant protein C deficiency. In the paper a case of interstitial lung disease in the course of the surfactant protein C deficiency coexisting with the primary humoral immunodeficiency is presented, with the discussion concerning clinical manifestation, radiological pattern, and histological findings.


Subject(s)
Immunity, Humoral , Immunologic Deficiency Syndromes/complications , Lung Diseases, Interstitial/diagnosis , Lung Diseases, Interstitial/therapy , Pulmonary Surfactant-Associated Protein C/deficiency , Child , Disease Susceptibility/immunology , Female , Humans , Lung Diseases, Interstitial/immunology
18.
Am J Physiol Lung Cell Mol Physiol ; 297(1): L64-72, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19304906

ABSTRACT

Patients with mutations in the pulmonary surfactant protein C (SP-C) gene develop interstitial lung disease and pulmonary exacerbations associated with viral infections including respiratory syncytial virus (RSV). Pulmonary infection with RSV caused more severe interstitial thickening, air space consolidation, and goblet cell hyperplasia in SP-C-deficient (Sftpc(-/-)) mice compared with SP-C replete mice. The RSV-induced pathology resolved more slowly in Sftpc(-/-) mice with lung inflammation persistent up to 30 days postinfection. Polymorphonuclear leukocyte and macrophage counts were increased in the bronchoalveolar lavage (BAL) fluid of Sftpc(-/-) mice. Viral titers and viral F and G protein mRNA were significantly increased in both Sftpc(-/-) and heterozygous Sftpc(+/-) mice compared with controls. Expression of Toll-like receptor 3 (TLR3) mRNA was increased in the lungs of Sftpc(-/-) mice relative to Sftpc(+/+) mice before and after RSV infection. Consistent with the increased TLR3 expression, BAL inflammatory cells were increased in the Sftpc(-/-) mice after exposure to a TLR3-specific ligand, poly(I:C). Preparations of purified SP-C and synthetic phospholipids blocked poly(I:C)-induced TLR3 signaling in vitro. SP-C deficiency increases the severity of RSV-induced pulmonary inflammation through regulation of TLR3 signaling.


Subject(s)
Pulmonary Surfactant-Associated Protein C/deficiency , Respiratory Syncytial Virus Infections/metabolism , Respiratory Syncytial Virus Infections/pathology , Animals , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/virology , Cell Count , Cell Line , Collectins/metabolism , Disease Models, Animal , Disease Susceptibility , Gene Expression Regulation, Viral , Goblet Cells/pathology , Goblet Cells/virology , Humans , Hypertrophy , Ligands , Lung/metabolism , Lung/pathology , Lung/virology , Mice , Pneumonia/complications , Pneumonia/pathology , Pneumonia/virology , Pulmonary Surfactant-Associated Protein C/metabolism , RNA, Double-Stranded/metabolism , Respiratory Syncytial Virus Infections/complications , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Viruses/genetics , Time Factors , Toll-Like Receptor 3/metabolism
19.
J Immunol ; 181(1): 621-8, 2008 Jul 01.
Article in English | MEDLINE | ID: mdl-18566429

ABSTRACT

To determine the role of surfactant protein C (SP-C) in host defense, SP-C-deficient (Sftpc-/-) mice were infected with the pulmonary pathogen Pseudomonas aeruginosa by intratracheal injection. Survival of young, postnatal day 14 Sftpc-/- mice was decreased in comparison to Sftpc+/+ mice. The sensitivity to Pseudomonas bacteria was specific to the 129S6 strain of Sftpc-/- mice, a strain that spontaneously develops interstitial lung disease-like lung pathology with age. Pulmonary bacterial load and leukocyte infiltration were increased in the lungs of Sftpc-/- mice 24 h after infection. Early influx of polymorphonuclear leukocytes in the lungs of uninfected newborn Sftpc-/- mice relative to Sftpc+/+ mice indicate that the lack of SP-C promotes proinflammatory responses in the lung. Mucin expression, as indicated by Alcian blue staining, was increased in the airways of Sftpc-/- mice following infection. Phagocytic activity of alveolar macrophages from Sftpc-/- mice was reduced. The uptake of fluorescent beads in vitro and the number of bacteria phagocytosed by alveolar macrophages in vivo was decreased in the Sftpc-/- mice. Alveolar macrophages from Sftpc-/- mice expressed markers of alternative activation that are associated with diminished pathogen response and advancing pulmonary fibrosis. These findings implicate SP-C as a modifier of alveolar homeostasis. SP-C plays an important role in innate host defense of the lung, enhancing macrophage-mediated Pseudomonas phagocytosis, clearance and limiting pulmonary inflammatory responses.


Subject(s)
Macrophages/immunology , Pseudomonas Infections/immunology , Pseudomonas Infections/metabolism , Pseudomonas aeruginosa/immunology , Pulmonary Surfactant-Associated Protein C/deficiency , Pulmonary Surfactant-Associated Protein C/immunology , Pulmonary Surfactant-Associated Protein C/metabolism , Animals , Biomarkers , Disease Models, Animal , Hyperplasia/genetics , Hyperplasia/metabolism , Hyperplasia/pathology , Mice , Mice, Knockout , Phagocytosis , Pseudomonas Infections/genetics , Pseudomonas Infections/pathology , Pulmonary Surfactant-Associated Protein C/genetics , Survival Rate
20.
Pediatr Res ; 63(6): 645-9, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18317237

ABSTRACT

The prevalence of the common mutations in the surfactant protein-B (121ins2), surfactant protein-C (I73T), and ATP-binding cassette member A3 (E292V) genes in population-based or case-control cohorts of newborn respiratory distress syndrome (RDS) is unknown. We determined the frequencies of these mutations in ethnically diverse population and disease-based cohorts using restriction enzyme analysis (121ins2 and E292V) and a 5' nuclease assay (I73T) in DNA samples from population-based cohorts in Missouri, Norway, South Korea, and South Africa, and from a case-control cohort of newborns with and without RDS (n = 420). We resequenced the ATP-binding cassette member A3 gene (ABCA3) in E292V carriers and computationally inferred ABCA3 haplotypes. The population-based frequencies of 121ins2, E292V, and I73T were rare (<0.4%). E292V was present in 3.8% of newborns with RDS, a 10-fold greater prevalence than in the Missouri cohort (p < 0.001). We did not identify other loss of function mutations in ABCA3 among patients with E292V that would account for their RDS. E292V occurred on a unique haplotype that was derived from a recombination of two common ABCA3 haplotypes. E292V was over-represented in newborns with RDS suggesting that E292V or its unique haplotype impart increased genetic risk for RDS.


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 , ATP-Binding Cassette Transporters/metabolism , Case-Control Studies , Female , Gene Frequency , Genetic Predisposition to Disease , Haplotypes , Humans , Infant , Infant, Newborn , Korea , Male , Missouri , Norway , Population Surveillance , Pulmonary Surfactant-Associated Protein B/deficiency , Pulmonary Surfactant-Associated Protein C/deficiency , Respiratory Distress Syndrome, Newborn/metabolism , Risk Factors , South Africa
SELECTION OF CITATIONS
SEARCH DETAIL
...