Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 142
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Cell ; 168(3): 460-472.e14, 2017 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-28089356

RESUMEN

Certain cell types function as factories, secreting large quantities of one or more proteins that are central to the physiology of the respective organ. Examples include surfactant proteins in lung alveoli, albumin in liver parenchyma, and lipase in the stomach lining. Whole-genome sequencing analysis of lung adenocarcinomas revealed noncoding somatic mutational hotspots near VMP1/MIR21 and indel hotspots in surfactant protein genes (SFTPA1, SFTPB, and SFTPC). Extrapolation to other solid cancers demonstrated highly recurrent and tumor-type-specific indel hotspots targeting the noncoding regions of highly expressed genes defining certain secretory cellular lineages: albumin (ALB) in liver carcinoma, gastric lipase (LIPF) in stomach carcinoma, and thyroglobulin (TG) in thyroid carcinoma. The sequence contexts of indels targeting lineage-defining genes were significantly enriched in the AATAATD DNA motif and specific chromatin contexts, including H3K27ac and H3K36me3. Our findings illuminate a prevalent and hitherto unrecognized mutational process linking cellular lineage and cancer.


Asunto(s)
Linaje de la Célula , Mutación INDEL , Mutación , Neoplasias/genética , Neoplasias/patología , Regiones no Traducidas 3' , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Masculino , Proteínas de la Membrana/genética , MicroARNs/genética , Persona de Mediana Edad , Motivos de Nucleótidos , Polimorfismo de Nucleótido Simple , Proteínas Asociadas a Surfactante Pulmonar/genética
2.
Proc Natl Acad Sci U S A ; 119(6)2022 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-35121658

RESUMEN

Pathogenic variants in surfactant proteins SP-B and SP-C cause surfactant deficiency and interstitial lung disease. Surfactant proteins are synthesized as precursors (proSP-B, proSP-C), trafficked, and processed via a vesicular-regulated secretion pathway; however, control of vesicular trafficking events is not fully understood. Through the Undiagnosed Diseases Network, we evaluated a child with interstitial lung disease suggestive of surfactant deficiency. Variants in known surfactant dysfunction disorder genes were not found in trio exome sequencing. Instead, a de novo heterozygous variant in RAB5B was identified in the Ras/Rab GTPases family nucleotide binding domain, p.Asp136His. Functional studies were performed in Caenorhabditis elegans by knocking the proband variant into the conserved position (Asp135) of the ortholog, rab-5 Genetic analysis demonstrated that rab-5[Asp135His] is damaging, producing a strong dominant negative gene product. rab-5[Asp135His] heterozygotes were also defective in endocytosis and early endosome (EE) fusion. Immunostaining studies of the proband's lung biopsy revealed that RAB5B and EE marker EEA1 were significantly reduced in alveolar type II cells and that mature SP-B and SP-C were significantly reduced, while proSP-B and proSP-C were normal. Furthermore, staining normal lung showed colocalization of RAB5B and EEA1 with proSP-B and proSP-C. These findings indicate that dominant negative-acting RAB5B Asp136His and EE dysfunction cause a defect in processing/trafficking to produce mature SP-B and SP-C, resulting in interstitial lung disease, and that RAB5B and EEs normally function in the surfactant secretion pathway. Together, the data suggest a noncanonical function for RAB5B and identify RAB5B p.Asp136His as a genetic mechanism for a surfactant dysfunction disorder.


Asunto(s)
Variación Genética/genética , Precursores de Proteínas/genética , Proteína C Asociada a Surfactante Pulmonar/genética , Proteínas Asociadas a Surfactante Pulmonar/genética , Proteínas de Unión al GTP rab5/genética , Células Epiteliales Alveolares/metabolismo , Animales , Caenorhabditis elegans/genética , Humanos , Pulmón/metabolismo , Enfermedades Pulmonares Intersticiales/genética , Surfactantes Pulmonares/metabolismo
3.
Eur Respir J ; 63(5)2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38575158

RESUMEN

BACKGROUND: Several rare surfactant-related gene (SRG) variants associated with interstitial lung disease are suspected to be associated with lung cancer, but data are missing. We aimed to study the epidemiology and phenotype of lung cancer in an international cohort of SRG variant carriers. METHODS: We conducted a cross-sectional study of all adults with SRG variants in the OrphaLung network and compared lung cancer risk with telomere-related gene (TRG) variant carriers. RESULTS: We identified 99 SRG adult variant carriers (SFTPA1 (n=18), SFTPA2 (n=31), SFTPC (n=24), ABCA3 (n=14) and NKX2-1 (n=12)), including 20 (20.2%) with lung cancer (SFTPA1 (n=7), SFTPA2 (n=8), SFTPC (n=3), NKX2-1 (n=2) and ABCA3 (n=0)). Among SRG variant carriers, the odds of lung cancer was associated with age (OR 1.04, 95% CI 1.01-1.08), smoking (OR 20.7, 95% CI 6.60-76.2) and SFTPA1/SFTPA2 variants (OR 3.97, 95% CI 1.39-13.2). Adenocarcinoma was the only histological type reported, with programmed death ligand-1 expression ≥1% in tumour cells in three samples. Cancer staging was localised (I/II) in eight (40%) individuals, locally advanced (III) in two (10%) and metastatic (IV) in 10 (50%). We found no somatic variant eligible for targeted therapy. Seven cancers were surgically removed, 10 received systemic therapy, and three received the best supportive care according to their stage and performance status. The median overall survival was 24 months, with stage I/II cancers showing better survival. We identified 233 TRG variant carriers. The comparative risk (subdistribution hazard ratio) for lung cancer in SRG patients versus TRG patients was 18.1 (95% CI 7.1-44.7). CONCLUSIONS: The high risk of lung cancer among SRG variant carriers suggests specific screening and diagnostic and therapeutic challenges. The benefit of regular computed tomography scan follow-up should be evaluated.


Asunto(s)
Neoplasias Pulmonares , Proteína A Asociada a Surfactante Pulmonar , Proteína C Asociada a Surfactante Pulmonar , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Masculino , Femenino , Persona de Mediana Edad , Anciano , Estudios Transversales , Proteína C Asociada a Surfactante Pulmonar/genética , Proteína A Asociada a Surfactante Pulmonar/genética , Adulto , Factor Nuclear Tiroideo 1/genética , Transportadoras de Casetes de Unión a ATP/genética , Factores de Riesgo , Predisposición Genética a la Enfermedad , Enfermedades Pulmonares Intersticiales/genética , Heterocigoto , Proteínas Asociadas a Surfactante Pulmonar/genética
4.
Mol Cell Proteomics ; 19(9): 1523-1532, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32581039

RESUMEN

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


Asunto(s)
Antiinfecciosos/metabolismo , Antiinfecciosos/farmacología , Iguanas/metabolismo , Sistema Inmunológico/metabolismo , Inmunidad Innata , Proteoma/metabolismo , Transcriptoma , Animales , Apoproteínas/genética , Apoproteínas/metabolismo , Bacillus subtilis/efectos de los fármacos , Encéfalo/metabolismo , Factores Quimiotácticos/genética , Factores Quimiotácticos/metabolismo , Ecuador , Endopeptidasas/genética , Endopeptidasas/metabolismo , Escherichia coli/efectos de los fármacos , Galectinas/genética , Galectinas/metabolismo , Corazón/fisiología , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Iguanas/genética , Iguanas/inmunología , Inmunidad Innata/genética , Pulmón/metabolismo , Muramidasa/genética , Muramidasa/metabolismo , Músculos/metabolismo , Miocardio/metabolismo , Especificidad de Órganos , Proteoma/genética , Proteoma/inmunología , Proteómica , Proteínas Asociadas a Surfactante Pulmonar/genética , Proteínas Asociadas a Surfactante Pulmonar/metabolismo , Piel/metabolismo , Espectrometría de Masas en Tándem , Transcriptoma/genética
5.
Am J Physiol Lung Cell Mol Physiol ; 320(5): L942-L957, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33719563

RESUMEN

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


Asunto(s)
Dexametasona/farmacología , Glucocorticoides/farmacología , Precursores de Proteínas/metabolismo , Proteínas Asociadas a Surfactante Pulmonar/metabolismo , Estabilidad del ARN/efectos de los fármacos , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , Células A549 , Animales , Células HEK293 , Humanos , Ratones , Precursores de Proteínas/genética , Proteínas Asociadas a Surfactante Pulmonar/genética , ARN Mensajero/genética , Proteínas de Unión al ARN/genética
6.
Protein Expr Purif ; 179: 105801, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33248225

RESUMEN

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


Asunto(s)
Antibacterianos , Proteínas Asociadas a Surfactante Pulmonar , Proteínas Recombinantes , Animales , Antibacterianos/metabolismo , Antibacterianos/farmacología , Bacterias/efectos de los fármacos , Escherichia coli/genética , Hongos/efectos de los fármacos , Pulmón/química , Pruebas de Sensibilidad Microbiana , Proteínas Asociadas a Surfactante Pulmonar/genética , Proteínas Asociadas a Surfactante Pulmonar/metabolismo , Proteínas Asociadas a Surfactante Pulmonar/farmacología , ARN/aislamiento & purificación , Ratas , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología
7.
J Cell Mol Med ; 23(11): 7664-7672, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31490024

RESUMEN

MicroRNA (miRNA) critically controls gene expression in many biological processes, including lung growth and pulmonary surfactant biosynthesis. The present study was conducted to investigate whether miR-20a-5p had such regulatory functions on alveolar type II (AT-II) cells. To accomplish this, miR-20a-5p-overexpressed and miR-20a-5p-inhibited adenoviral vectors were constructed and transfected into cultured AT-II cells that were isolated from rat foetal lungs of 19 days' gestation. Transfection efficiency was confirmed by observing the fluorescence of green fluorescent protein (GFP) carried by the viral vector, whereas miR-20a-5p levels were verified by real-time PCR. The CCK-8 assay was used to compare the proliferation ability of AT-II cells that had over- or underexpressed miR-20a-5p. The expression of surfactant-associated proteins (SPs) and phosphatase and tensin homolog (PTEN) was measured by real-time PCR and Western blotting. In AT-II cells, transfection resulted in over- or under-regulation of miR-20a-5p. While overexpression of miR-20a-5p promoted pulmonary surfactant gene expression, its underexpression inhibited it. Consistent with its role in negatively regulating the pulmonary surfactant gene, an opposite pattern was observed for miR-20a-5p regulation of PTEN. As a result, when miR-20a-5p was rendered overexpressed, PTEN was down-regulated. By contrast, when miR-20a-5p was underexpressed, PTEN was up-regulated. Neither overexpression nor underexpression of miR-20a-5p altered the cell proliferation. miR-20a-5p plays no role in proliferation of foetal AT-II cells but is a critical regulator of surfactant gene expression. The latter appears to be achieved through a regulatory process that implicates expression of PTEN.


Asunto(s)
Células Epiteliales Alveolares/metabolismo , Regulación de la Expresión Génica , MicroARNs/metabolismo , Proteínas Asociadas a Surfactante Pulmonar/genética , Células Epiteliales Alveolares/citología , Animales , Secuencia de Bases , Proliferación Celular/genética , Análisis por Conglomerados , Regulación hacia Abajo/genética , Humanos , Recién Nacido , MicroARNs/genética , Fosfohidrolasa PTEN/metabolismo , Proteínas Asociadas a Surfactante Pulmonar/metabolismo , Ratas Sprague-Dawley , Regulación hacia Arriba/genética
8.
Biochem Biophys Res Commun ; 499(4): 843-848, 2018 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-29621540

RESUMEN

TGF beta is a multifunctional cytokine that regulates alveolar epithelial cells as well as immune cells and fibroblasts. TGF beta inhibits surfactant protein A, B and C expression in fetal human lung and can inhibit type II cell proliferation induced by FGF7 (KGF). However, little is known about direct effects of TGF beta on adult human type II cells. We cultured alveolar type II cells under air/liquid interface conditions to maintain their state of differentiation with or without TGF beta. TGF beta markedly decreased expression of SP-A, SP-B, SP-C, fatty acid synthase, and the phospholipid transporter ABCA3. However, TGF beta increased protein levels of SP-D with little change in mRNA levels, indicating that it is regulated independently from other components of surfactant. TGF beta is a negative regulator of both the protein and the phospholipid components of surfactant. TGF beta did not induce EMT changes in highly differentiated human type II cells. SP-D is an important host defense molecule and regulated independently from the other surfactant proteins. Taken together these data are the first report of the effect of TGF beta on highly differentiated adult human type II cells. The effects on the surfactant system are likely important in the development of fibrotic lung diseases.


Asunto(s)
Células Epiteliales Alveolares/metabolismo , Proteínas Asociadas a Surfactante Pulmonar/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Células Epiteliales Alveolares/efectos de los fármacos , Animales , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Lipogénesis/efectos de los fármacos , Proteínas Asociadas a Surfactante Pulmonar/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas
9.
Am J Obstet Gynecol ; 218(1): 132.e1-132.e9, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29138038

RESUMEN

BACKGROUND: Antenatal steroids are standard of care for women who are at risk of preterm delivery; however, antenatal steroid dosing and formulation have not been evaluated adequately. The standard clinical 2-dose treatment with betamethasone-acetate+betamethasone-phosphate is more effective than 2 doses of betamethasone-phosphate for the induction of lung maturation in preterm fetal sheep. We hypothesized that the slowly released betamethasone-acetate component induces similar lung maturation to betamethasone-phosphate+betamethasone-acetate with decreased dose and fetal exposure. OBJECTIVE: The purpose of this study was to investigate pharmacokinetics and fetal lung maturation of antenatal betamethasone-acetate in preterm fetal sheep. STUDY DESIGN: Groups of 10 singleton-pregnant ewes received 1 or 2 intramuscular doses 24 hours apart of 0.25 mg/kg/dose of betamethasone-phosphate+betamethasone-acetate (the standard of care dose) or 1 intramuscular dose of 0.5 mg/kg, 0.25 mg/kg, or 0.125 mg/kg of betamethasone-acetate. Fetuses were delivered 48 hours after the first injection at 122 days of gestation (80% of term) and ventilated for 30 minutes, with ventilator settings, compliance, vital signs, and blood gas measurements recorded every 10 minutes. After ventilation, we measured static lung pressure-volume curves and sampled the lungs for messenger RNA measurements. Other groups of pregnant ewes and fetuses were catheterized and treated with intramuscular injections of betamethasone-phosphate 0.125 mg/kg, betamethasone-acetate 0.125 mg/kg, or betamethasone-acetate 0.5 mg/kg. Maternal and fetal betamethasone concentrations in plasma were measured for 24 hours. RESULTS: All betamethasone-treated groups had increased messenger RNA expression of surfactant proteins A, B, and C, ATP-binding cassette subfamily A member 3, and aquaporin-5 compared with control animals. Treatment with 1 dose of intramuscular betamethasone-acetate 0.125mg/kg improved dynamic and static lung compliance, gas exchange, and ventilation efficiency similarly to the standard treatment of 2 doses of 0.25 m/kg of betamethasone-acetate+betamethasone-phosphate. Betamethasone-acetate 0.125 mg/kg resulted in lower maternal and fetal peak plasma concentrations and decreased fetal exposure to betamethasone compared with betamethasone-phosphate 0.125 mg/kg. CONCLUSION: A single dose of betamethasone-acetate results in similar fetal lung maturation as the 2-dose clinical formulation of betamethasone-phosphate+betamethasone-acetate with decreased fetal exposure to betamethasone. A lower dose of betamethasone-acetate may be an effective alternative to induce fetal lung maturation with less risk to the fetus.


Asunto(s)
Betametasona/administración & dosificación , Madurez de los Órganos Fetales/efectos de los fármacos , Glucocorticoides/administración & dosificación , Pulmón/efectos de los fármacos , Subfamilia A de Transportadores de Casete de Unión al ATP/genética , Subfamilia A de Transportadores de Casete de Unión al ATP/metabolismo , Animales , Acuaporina 5/genética , Acuaporina 5/metabolismo , Betametasona/análogos & derivados , Betametasona/farmacocinética , Relación Dosis-Respuesta a Droga , Femenino , Glucocorticoides/farmacocinética , Modelos Animales , Embarazo , Proteínas Asociadas a Surfactante Pulmonar/genética , Proteínas Asociadas a Surfactante Pulmonar/metabolismo , ARN Mensajero/metabolismo , Ovinos
10.
Curr Opin Pulm Med ; 24(3): 253-259, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29517585

RESUMEN

PURPOSE OF REVIEW: Interstitial lung disease (ILD) in children (chILD) is an umbrella term for a heterogeneous group of rare respiratory disorders that are mostly chronic and associated with high morbidity and mortality. The pathogenesis of the various chILD is complex and implicates genetic contributors. The purpose of this review is to provide updated information on the molecular defects associated with the development of chILD. RECENT FINDINGS: Currently, the main mutations are identified in the surfactant genes SFTPA1, SFTPA2, SFTPB, SFTPC, ABCA3, and NKX2-1. In addition, pulmonary alveolar proteinosis is associated with mutations in CSF2RA, CSF2RB, and MARS, and specific auto-inflammatory forms of chILD implicate STING and COPA disorders. The relationships between the genetic defects and the disease expression remain poorly understood, with no genotype-phenotype correlations identified so far. Although targeted therapies are emerging, the management strategies are still largely empirical, relying mostly on corticosteroids. SUMMARY: Genetic factors play an important role in chILD, and the ongoing development of novel technologies will rapidly broaden the genetic landscape of chILD. Therefore, in the coming years, it is expected that newly identified molecular defects and markers will help predicting disease courses and tailoring individual therapies.


Asunto(s)
Enfermedades Pulmonares Intersticiales/genética , Proteínas Asociadas a Surfactante Pulmonar/genética , Transportadoras de Casetes de Unión a ATP/genética , Niño , Asesoramiento Genético , Pruebas Genéticas , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Humanos , Enfermedades Pulmonares Intersticiales/terapia , Mutación , Fenotipo , Factor Nuclear Tiroideo 1/genética
11.
Pulm Pharmacol Ther ; 52: 18-26, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30130572

RESUMEN

BACKGROUND: Dexamethasone (DEX) is widely used for antenatal lung maturation and has been investigated to prevent premature lung injury by inhibiting postnatal inflammation. Its pharmacological mechanisms in the treatment of bacterial infection-induced injury of neonatal lung parenchymal cells remain to be clarified. We hypothesized that DEX pretreatment may attenuate endotoxin-induced growth suppression and regulate cytokine mRNA expression in cultured neonatal type II alveolar epithelial cells (AEC-II). METHODS: AEC-II of newborn piglets were freshly isolated and cultured. After pretreatment of 0.01, 0.1, 1.0 and 10 µmol/l DEX (E0.01, E0.1, E1.0 and E10 group, respectively) for 24 h, the cells were cultured with 1 µg/ml lipopolysaccharides (LPS) for 7 days with medium replacement every 24 h. Messenger RNA expression of surfactant proteins (SPs), pro-inflammatory cytokines and multiple growth factors (GF) were determined by RT-PCR, along with the cell growth and apoptosis measurements. RESULTS: LPS without DEX pretreatment suppressed cell proliferation, enhanced expression of pro-inflammatory cytokine mRNA and apoptosis, which was ameliorated in all DEX-pretreated groups on day 3. On day 3 and 5, only cells pretreated by E1.0 and E10 showed a 20-fold increase in insulin-like GF-1 mRNA expression whereas the expression of other GFs was down-regulated. LPS exposure reduced the expression of SP-A, B, C and Aquaporin-5 mRNA on day 3-7. However, the expression of SP-C mRNA was increased in E1.0 on day 3, which was supported by in situ expression of pro-SP-C with immunocytochemical assay. CONCLUSION: LPS-induced in vitro AEC-II injury was partially prevented by DEX pretreatment, with 1.0 µmol/l being the potentially optimal concentration. (253 words).


Asunto(s)
Células Epiteliales Alveolares/efectos de los fármacos , Dexametasona/farmacología , Lipopolisacáridos/farmacología , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/patología , Animales , Apoptosis/efectos de los fármacos , Células Cultivadas , Citocinas/biosíntesis , Citocinas/genética , Relación Dosis-Respuesta a Droga , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Péptidos y Proteínas de Señalización Intercelular/genética , Masculino , Proteínas Asociadas a Surfactante Pulmonar/biosíntesis , Proteínas Asociadas a Surfactante Pulmonar/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Mensajero/metabolismo , Porcinos
12.
J Physiol ; 595(21): 6635-6652, 2017 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-28759122

RESUMEN

KEY POINTS: Offspring of overweight and obese women are at greater risk for respiratory complications at birth. We determined the effect of late gestation maternal overnutrition (LGON) in sheep on surfactant maturation, glucose transport and fatty acid metabolism in the lung in fetal and postnatal life. There were significant decreases in surfactant components and numerical density of surfactant producing cells in the alveolar epithelium due to LGON in the fetal lung. However, there were no differences in the levels of these surfactant components between control and LGON lambs at 30 days of age. The reduced capacity for surfactant production in fetuses as a result of LGON may affect the transition to air breathing at birth. There was altered glucose transport and fatty acid metabolism in the lung as a result of LGON in postnatal life. However, there is a normalisation of surfactant components that suggests accelerated maturation in the lungs after birth. ABSTRACT: With the increasing incidence of obesity worldwide, the proportion of women entering pregnancy overweight or obese has increased dramatically. The fetus of an overnourished mother experiences numerous metabolic changes that may modulate lung development and hence successful transition to air breathing at birth. We used a sheep model of maternal late gestation overnutrition (LGON; from 115 days' gestation, term 147 ± 3 days) to determine the effect of exposure to an increased plane of nutrition in late gestation on lung development in the fetus (at 141 days' gestation) and the lamb (30 days after birth). We found a decrease in the numerical density of surfactant protein positive cells, as well as a reduction in mRNA expression of surfactant proteins (SFTP-A, -B and -C), a rate limiting enzyme in surfactant phospholipid synthesis (phosphate cytidylyltransferase 1, choline, α; PCYT1A), and glucose transporters (SLC2A1 and SLC2A4) in the fetal lung. In lambs at 30 days after birth, there were no differences between Control and LGON groups in the surfactant components that were downregulated in the LGON fetuses. However, mRNA expression of SFTP-A, PCYT1A, peroxisome proliferator activated receptor-γ, fatty acid synthase and fatty acid transport protein were increased in LGON lambs compared to controls. These results indicate a reduced capacity for surfactant production in late gestation. While these deficits are normalised by 30 days after birth, the lungs of LGON lambs exhibited altered glucose transport and fatty acid metabolism, which is consistent with an enhanced capacity for surfactant synthesis and restoration of surfactant maturity in these animals.


Asunto(s)
Pulmón/embriología , Hipernutrición/metabolismo , Complicaciones del Embarazo/metabolismo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Proteínas Asociadas a Surfactante Pulmonar/metabolismo , Animales , Ácidos Grasos/metabolismo , Femenino , Glucosa/metabolismo , Pulmón/metabolismo , Pulmón/patología , Hipernutrición/patología , Embarazo , Complicaciones del Embarazo/patología , Efectos Tardíos de la Exposición Prenatal/patología , Proteínas Asociadas a Surfactante Pulmonar/genética , Mucosa Respiratoria/embriología , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/patología , Ovinos
13.
J Physiol ; 595(13): 4329-4350, 2017 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-28318025

RESUMEN

KEY POINTS: Chronic fetal hypoxaemia is a common pregnancy complication associated with intrauterine growth restriction that may influence respiratory outcome at birth. We investigated the effect of maternal chronic hypoxia for a month in late gestation on signalling pathways regulating fetal lung maturation and the transition to air-breathing at birth using isobaric hypoxic chambers without alterations to maternal food intake. Maternal chronic hypoxia in late gestation increases fetal lung expression of genes regulating hypoxia signalling, lung liquid reabsorption and surfactant maturation, which may be an adaptive response in preparation for the successful transition to air-breathing at birth. In contrast to other models of chronic fetal hypoxaemia, late gestation onset fetal hypoxaemia promotes molecular regulation of fetal lung maturation. This suggests a differential effect of timing and duration of fetal chronic hypoxaemia on fetal lung maturation, which supports the heterogeneity observed in respiratory outcomes in newborns following exposure to chronic hypoxaemia in utero. ABSTRACT: Chronic fetal hypoxaemia is a common pregnancy complication that may arise from maternal, placental and/or fetal factors. Respiratory outcome of the infant at birth likely depends on the duration, timing and severity of the hypoxaemic insult. We have isolated the effect of maternal chronic hypoxia (MCH) for a month in late gestation on fetal lung development. Pregnant ewes were exposed to normoxia (21% O2 ) or hypoxia (10% O2 ) from 105 to 138 days of gestation (term ∼145 days). At 138 days, gene expression in fetal lung tissue was determined by quantitative RT-PCR. Cortisol concentrations were determined in fetal plasma and lung tissue. Numerical density of surfactant protein positive cells was determined by immunohistochemistry. MCH reduced maternal PaO2 (106 ± 2.9 vs. 47 ± 2.8 mmHg) and fetal body weight (4.0 ± 0.4 vs. 3.2 ± 0.9 kg). MCH increased fetal lung expression of the anti-oxidant marker CAT and decreased expression of the pro-oxidant marker NOX-4. MCH increased expression of genes regulating hypoxia signalling and feedback (HIF-3α, KDM3A, SLC2A1, EGLN-3). There was no effect of MCH on fetal plasma/lung tissue cortisol concentrations, nor genes regulating glucocorticoid signalling (HSD11B-1, HSD11B-2, NR3C1, NR3C2). MCH increased expression of genes regulating sodium (SCNN1-B, ATP1-A1, ATP1-B1) and water (AQP-4) movement in the fetal lung. MCH promoted surfactant maturation (SFTP-B, SFTP-D, ABCA3) at the molecular level, but did not alter the numerical density of surfactant positive cells in lung tissue. MCH in late gestation promotes molecular maturation of the fetal lung, which may be an adaptive response in preparation for the successful transition to air-breathing at birth.


Asunto(s)
Hipoxia Fetal/metabolismo , Prolina Dioxigenasas del Factor Inducible por Hipoxia/metabolismo , Pulmón/metabolismo , Proteínas Asociadas a Surfactante Pulmonar/genética , 11-beta-Hidroxiesteroide Deshidrogenasas/genética , 11-beta-Hidroxiesteroide Deshidrogenasas/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Femenino , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/metabolismo , Prolina Dioxigenasas del Factor Inducible por Hipoxia/genética , Histona Demetilasas con Dominio de Jumonji/genética , Histona Demetilasas con Dominio de Jumonji/metabolismo , Pulmón/embriología , Pulmón/fisiología , Masculino , Embarazo , Proteínas Asociadas a Surfactante Pulmonar/metabolismo , Ovinos
14.
Am J Physiol Lung Cell Mol Physiol ; 313(6): L1030-L1046, 2017 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-28839101

RESUMEN

We previously proposed a role for the two-pore domain potassium (K2P) channel TREK-1 in hyperoxia (HO)-induced lung injury. To determine whether redundancy among the three TREK isoforms (TREK-1, TREK-2, and TRAAK) could protect from HO-induced injury, we now examined the effect of deletion of all three TREK isoforms in a clinically relevant scenario of prolonged HO exposure and mechanical ventilation (MV). We exposed WT and TREK-1/TREK-2/TRAAK-deficient [triple knockout (KO)] mice to either room air, 72-h HO, MV [high and low tidal volume (TV)], or a combination of HO + MV and measured quasistatic lung compliance, bronchoalveolar lavage (BAL) protein concentration, histologic lung injury scores (LIS), cellular apoptosis, and cytokine levels. We determined surfactant gene and protein expression and attempted to prevent HO-induced lung injury by prophylactically administering an exogenous surfactant (Curosurf). HO treatment increased lung injury in triple KO but not WT mice, including an elevated LIS, BAL protein concentration, and markers of apoptosis, decreased lung compliance, and a more proinflammatory cytokine phenotype. MV alone had no effect on lung injury markers. Exposure to HO + MV (low TV) further decreased lung compliance in triple KO but not WT mice, and HO + MV (high TV) was lethal for triple KO mice. In triple KO mice, the HO-induced lung injury was associated with decreased surfactant protein (SP) A and SPC but not SPB and SPD expression. However, these changes could not be explained by alterations in the transcription factors nuclear factor-1 (NF-1), NKX2.1/thyroid transcription factor-1 (TTF-1) or c-jun, or lamellar body levels. Prophylactic Curosurf administration did not improve lung injury scores or compliance in triple KO mice.


Asunto(s)
Hiperoxia/metabolismo , Lesión Pulmonar/metabolismo , Canales de Potasio de Dominio Poro en Tándem/deficiencia , Canales de Potasio/deficiencia , Proteínas Asociadas a Surfactante Pulmonar/biosíntesis , Animales , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Hiperoxia/genética , Hiperoxia/patología , Lipopolisacáridos/toxicidad , Lesión Pulmonar/genética , Lesión Pulmonar/patología , Ratones , Ratones Noqueados , Proteínas Asociadas a Surfactante Pulmonar/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
15.
J Immunol ; 195(4): 1628-36, 2015 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-26163587

RESUMEN

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.


Asunto(s)
Proteínas Asociadas a Surfactante Pulmonar/metabolismo , Animales , Antibacterianos/farmacología , Péptidos Catiónicos Antimicrobianos/farmacología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Humanos , Concentración de Iones de Hidrógeno , Infecciones por Klebsiella/inmunología , Infecciones por Klebsiella/metabolismo , Infecciones por Klebsiella/microbiología , Klebsiella pneumoniae/efectos de los fármacos , Klebsiella pneumoniae/inmunología , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/microbiología , Pulmón/patología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Noqueados , Infiltración Neutrófila , Unión Proteica , Proteína A Asociada a Surfactante Pulmonar/genética , Proteína A Asociada a Surfactante Pulmonar/metabolismo , Proteína A Asociada a Surfactante Pulmonar/farmacología , Proteína B Asociada a Surfactante Pulmonar/genética , Proteína B Asociada a Surfactante Pulmonar/metabolismo , Proteína B Asociada a Surfactante Pulmonar/farmacología , Proteínas Asociadas a Surfactante Pulmonar/genética , Proteínas Asociadas a Surfactante Pulmonar/farmacología , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología
16.
Genet Mol Res ; 16(3)2017 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-28873200

RESUMEN

As a ß2 integrin family member, Mac-1 plays an important role in the inflammatory response. Inflammation and lung injury are closely associated, but the involvement of Mac-1 in the occurrence and development of such pathologies remains poorly understood. We aimed to investigate the relationship between Mac-1 deficiency and respiratory failure in Mac-1 knockout {Mac-1-/-} mice, using C57BL/6J mice as a control. The newborn survival rate of Mac-1-/- mice was calculated, and mouse lung tissue was treated with hematoxylin and eosin and subjected to immunofluorescent staining. Moreover, western blotting and immunohistochemistry were used to detect the expression of molecules specific to type I and type II alveolar epithelial cells, as well as alveolar surfactant proteins secreted by the latter. Survival of Mac-1-/- pups was significantly lower than that of newborn C57BL/6J mice. In a float test, lung tissues from C57BL/6J mice were buoyant, whereas those of Mac-1-/- mice were not. Compared with C57BL/6J mice, expression of proSP-C {specific to type II alveolar epithelial cells} and alveolar surfactant proteins in Mac-1-/- mice was not significantly different, implying that type II cell function was unaltered. However, western blotting revealed expression of T1α, Aqp5, and Snx5 {type I alveolar epithelial cell markers} in Mac-1-/- mice to be significantly decreased {P < 0.05}. In conclusion, Mac-1 may play an important role in respiratory failure. Its absence leads to this condition not by influencing type II alveolar epithelial cells or their secreted surfactant proteins, but rather by reducing type I alveolar cell numbers.


Asunto(s)
Células Epiteliales Alveolares/metabolismo , Antígeno CD11b/genética , Insuficiencia Respiratoria/metabolismo , Animales , Antígeno CD11b/metabolismo , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Asociadas a Surfactante Pulmonar/genética , Proteínas Asociadas a Surfactante Pulmonar/metabolismo , Insuficiencia Respiratoria/genética , Insuficiencia Respiratoria/patología
17.
Inhal Toxicol ; 28(9): 421-8, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27308969

RESUMEN

Occupational inhalation of indium compounds can cause the so-called "indium lung disease". Most affected individuals show pulmonary alveolar proteinosis (PAP) and fibrotic interstitial lung disease. In animal experiments, inhalation of indium tin oxide or indium oxide has been shown to cause lung damage. However, the mechanisms by which indium compounds lead to indium lung disease remain unknown. In this study, we constructed a mouse model of indium lung disease and analyzed gene expression in response to indium exposure. Indium oxide (In2O3, 10 mg/kg, primary particle size <100 nm) was administered intratracheally to C57BL/6 mice (male, 8 weeks of age) twice a week for 8 weeks. Four weeks after the final instillation, histopathological analysis exhibited periodic acid-Schiff positive material in the alveoli, characteristic of PAP. Comprehensive gene expression analysis by RNA-Seq, however, revealed expression of fibrosis-related genes, such as surfactant associated protein D, surfactant associated protein A1, mucin 1, and collagen type I and III, was significantly increased, indicating that fibrotic gene expression progresses in early phase of indium lung. These data supported the latest hypothesis that PAP occurs as an acute phase response and is replaced by fibrosis after long-term latency.


Asunto(s)
Expresión Génica/efectos de los fármacos , Indio/toxicidad , Exposición por Inhalación/efectos adversos , Enfermedades Pulmonares Intersticiales/inducido químicamente , Proteinosis Alveolar Pulmonar/inducido químicamente , Alveolos Pulmonares , Animales , Colágeno Tipo I/genética , Colágeno Tipo III/genética , Modelos Animales de Enfermedad , Fibrosis , Enfermedades Pulmonares Intersticiales/patología , Ratones Endogámicos C57BL , Mucina-1/genética , Tamaño de la Partícula , Proteinosis Alveolar Pulmonar/patología , Alveolos Pulmonares/efectos de los fármacos , Alveolos Pulmonares/patología , Proteínas Asociadas a Surfactante Pulmonar/genética
18.
Am J Physiol Lung Cell Mol Physiol ; 309(6): L507-25, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26186947

RESUMEN

Dating back nearly 35 years ago to the Witschi hypothesis, epithelial cell dysfunction and abnormal wound healing have reemerged as central concepts in the pathophysiology of idiopathic pulmonary fibrosis (IPF) in adults and in interstitial lung disease in children. Alveolar type 2 (AT2) cells represent a metabolically active compartment in the distal air spaces responsible for pulmonary surfactant biosynthesis and function as a progenitor population required for maintenance of alveolar integrity. Rare mutations in surfactant system components have provided new clues to understanding broader questions regarding the role of AT2 cell dysfunction in the pathophysiology of fibrotic lung diseases. Drawing on data generated from a variety of model systems expressing disease-related surfactant component mutations [surfactant proteins A and C (SP-A and SP-C); the lipid transporter ABCA3], this review will examine the concept of epithelial dysfunction in fibrotic lung disease, provide an update on AT2 cell and surfactant biology, summarize cellular responses to mutant surfactant components [including endoplasmic reticulum (ER) stress, mitochondrial dysfunction, and intrinsic apoptosis], and examine quality control pathways (unfolded protein response, the ubiquitin-proteasome system, macroautophagy) that can be utilized to restore AT2 homeostasis. This integrated response and its derangement will be placed in the context of cell stress and quality control signatures found in patients with familial or sporadic IPF as well as non-surfactant-related AT2 cell dysfunction syndromes associated with a fibrotic lung phenotype. Finally, the need for targeted therapeutic strategies for pulmonary fibrosis that address epithelial ER stress, its downstream signaling, and cell quality control are discussed.


Asunto(s)
Células Epiteliales Alveolares/fisiología , Fibrosis Pulmonar/metabolismo , Proteínas Asociadas a Surfactante Pulmonar/metabolismo , Animales , Apoptosis , Humanos , Mutación , Procesamiento Proteico-Postraduccional , Transporte de Proteínas , Alveolos Pulmonares/metabolismo , Alveolos Pulmonares/patología , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/patología , Proteínas Asociadas a Surfactante Pulmonar/genética , Transducción de Señal
19.
Clin Immunol ; 215: 108426, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32311462

Asunto(s)
Betacoronavirus/inmunología , Infecciones por Coronavirus/inmunología , Pneumocystis carinii/inmunología , Neumonía por Pneumocystis/inmunología , Neumonía Viral/inmunología , Proteínas Asociadas a Surfactante Pulmonar/química , Surfactantes Pulmonares/química , Glicoproteína de la Espiga del Coronavirus/química , Secuencia de Aminoácidos , Anticuerpos Antivirales/química , Anticuerpos Antivirales/genética , Betacoronavirus/patogenicidad , COVID-19 , Coronavirus Humano 229E/inmunología , Infecciones por Coronavirus/genética , Infecciones por Coronavirus/patología , Infecciones por Coronavirus/virología , Coronavirus Humano OC43/inmunología , Reacciones Cruzadas , Epítopos/química , Epítopos/genética , Epítopos/inmunología , Expresión Génica , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Oligopéptidos/química , Oligopéptidos/genética , Oligopéptidos/inmunología , Pandemias , Pneumocystis carinii/patogenicidad , Neumonía por Pneumocystis/genética , Neumonía por Pneumocystis/patología , Neumonía por Pneumocystis/virología , Neumonía Viral/genética , Neumonía Viral/patología , Neumonía Viral/virología , Unión Proteica , Proteínas Asociadas a Surfactante Pulmonar/genética , Proteínas Asociadas a Surfactante Pulmonar/inmunología , Surfactantes Pulmonares/inmunología , Surfactantes Pulmonares/metabolismo , SARS-CoV-2 , Homología de Secuencia de Aminoácido , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/inmunología
20.
Clin Immunol ; 217: 108480, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32461193

Asunto(s)
Autoanticuerpos/inmunología , Autoantígenos/inmunología , Enfermedades Autoinmunes/epidemiología , Betacoronavirus/inmunología , Infecciones por Coronavirus/epidemiología , Proteínas de la Nucleocápside/inmunología , Pandemias , Neumonía Viral/epidemiología , Glicoproteína de la Espiga del Coronavirus/inmunología , Anticuerpos Antivirales/química , Anticuerpos Antivirales/genética , Autoanticuerpos/química , Autoanticuerpos/genética , Autoantígenos/química , Autoantígenos/genética , Enfermedades Autoinmunes/etiología , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/virología , Autoinmunidad , Betacoronavirus/patogenicidad , COVID-19 , Vacunas contra la COVID-19 , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/prevención & control , Infecciones por Coronavirus/virología , Proteínas de la Nucleocápside de Coronavirus , Reacciones Cruzadas , Expresión Génica , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad Innata , Proteínas de la Nucleocápside/química , Proteínas de la Nucleocápside/genética , Pandemias/prevención & control , Fosfoproteínas , Neumonía Viral/inmunología , Neumonía Viral/prevención & control , Neumonía Viral/virología , Proteínas Asociadas a Surfactante Pulmonar/antagonistas & inhibidores , Proteínas Asociadas a Surfactante Pulmonar/genética , Proteínas Asociadas a Surfactante Pulmonar/inmunología , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/genética , Vacunas Virales/efectos adversos , Vacunas Virales/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA