Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell Mol Biol Lett ; 22: 23, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29167681

RESUMEN

BACKGROUND: Hypoxic conditions induce the expression of hypoxia-inducible factors (HIFs) that allow cells to adapt to the changing conditions and alter the expression of a number of genes including the cystic fibrosis transmembrane conductance regulator (CFTR). CFTR is a low abundance mRNA in airway epithelial cells even during normoxic conditions, but during hypoxia its mRNA expression decreases even further. METHODS: In the current studies, we examined the kinetics of hypoxia-induced changes in CFTR mRNA and protein levels in two human airway epithelial cell lines, Calu-3 and 16HBE14o-, and in normal primary bronchial epithelial cells. Our goal was to examine the posttranscriptional modifications that affected CFTR expression during hypoxia. We utilized in silico predictive protocols to establish potential miRNAs that could potentially regulate CFTR message stability and identified miR-200b as a candidate molecule. RESULTS: Analysis of each of the epithelial cell types during prolonged hypoxia revealed that CFTR expression decreased after 12 h during a time when miR-200b was continuously upregulated. Furthermore, manipulation of the miRNA levels during normoxia and hypoxia using miR-200b mimics and antagomirs decreased and increased CFTR mRNA levels, respectively, and thus established that miR-200b downregulates CFTR message levels during hypoxic conditions. CONCLUSION: The data suggest that miR-200b may be a suitable target for modulating CFTR levels in vivo.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Regulación hacia Abajo/genética , Células Epiteliales/metabolismo , Pulmón/citología , MicroARNs/metabolismo , Regiones no Traducidas 3'/genética , Regiones no Traducidas 5'/genética , Secuencia de Bases , Hipoxia de la Célula , Línea Celular , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , MicroARNs/genética , Modelos Biológicos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transcripción Genética
2.
Am J Physiol Lung Cell Mol Physiol ; 305(8): L530-41, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-23934925

RESUMEN

Chronic obstructive pulmonary disease (COPD) is a progressive respiratory disorder consisting of chronic bronchitis and/or emphysema. COPD patients suffer from chronic infections and display exaggerated inflammatory responses and a progressive decline in respiratory function. The respiratory symptoms of COPD are similar to those seen in cystic fibrosis (CF), although the molecular basis of the two disorders differs. CF is a genetic disease caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene encoding a chloride and bicarbonate channel (CFTR), leading to CFTR dysfunction. The majority of COPD cases result from chronic oxidative insults such as cigarette smoke. Interestingly, environmental stresses including cigarette smoke, hypoxia, and chronic inflammation have also been implicated in reduced CFTR function, and this suggests a common mechanism that may contribute to both the CF and COPD. Therefore, improving CFTR function may offer an excellent opportunity for the development of a common treatment for CF and COPD. In this article, we review what is known about the CF respiratory phenotype and discuss how diminished CFTR expression-associated ion transport defects may contribute to some of the pathological changes seen in COPD.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Regulación de la Expresión Génica , Estrés Oxidativo , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Fumar/efectos adversos , Fumar/metabolismo , Enfermedad Crónica , Fibrosis Quística/metabolismo , Fibrosis Quística/patología , Fibrosis Quística/terapia , Humanos , Inflamación , Transporte Iónico , Enfermedad Pulmonar Obstructiva Crónica/etiología , Enfermedad Pulmonar Obstructiva Crónica/patología , Enfermedad Pulmonar Obstructiva Crónica/terapia , Fumar/patología , Fumar/terapia
3.
Cell Mol Biol Lett ; 18(1): 47-57, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23124858

RESUMEN

Understanding the cellular pathways that regulate angiogenesis during hypoxia is a necessary aspect in the development of novel treatments for cardiovascular disorders. Although the pathways of angiogenesis have been extensively studied, there is limited information on the role of miRNAs in this process. miRNAs or their antagomirs could be used in future therapeutic approaches to regulate hypoxia-induced angiogenesis, so it is critical to understand their role in governing angiogenesis during hypoxic conditions. Although hypoxia and ischemia change the expression profile of many miRNAs, a functional role for a limited number of so-called hypoxamiRs has been demonstrated in angiogenesis. Here, we discuss the best examples that illustrate the role of hypoxamiRs in angiogenesis.


Asunto(s)
Enfermedades Cardiovasculares/genética , Regulación de la Expresión Génica , Hipoxia/genética , MicroARNs/genética , Neoplasias/irrigación sanguínea , Neovascularización Patológica , Regiones no Traducidas 3'/genética , Enfermedades Cardiovasculares/metabolismo , Hipoxia de la Célula , Línea Celular Tumoral , Células Endoteliales/metabolismo , Células Endoteliales/patología , Humanos , Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , MicroARNs/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
4.
Biochem J ; 441(2): 633-43, 2012 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-21995445

RESUMEN

CFTR (cystic fibrosis transmembrane conductance regulator) is expressed in the apical membrane of epithelial cells. Cell-surface CFTR levels are regulated by endocytosis and recycling. A number of adaptor proteins including AP-2 (µ2 subunit) and Dab2 (Disabled-2) have been proposed to modulate CFTR internalization. In the present study we have used siRNA (small interfering RNA)-mediated silencing of these adaptors to test their roles in the regulation of CFTR cell-surface trafficking and stability in human airway epithelial cells. The results indicate that µ2 and Dab2 performed partially overlapping, but divergent, functions. While µ2 depletion dramatically decreased CFTR endocytosis with little effect on the half-life of the CFTR protein, Dab2 depletion increased the CFTR half-life ~3-fold, in addition to inhibiting CFTR endocytosis. Furthermore, Dab2 depletion inhibited CFTR trafficking from the sorting endosome to the recycling compartment, as well as delivery of CFTR to the late endosome, thus providing a mechanistic explanation for increased CFTR expression and half-life. To test whether two E3 ligases were required for the endocytosis and/or down-regulation of surface CFTR, we siRNA-depleted CHIP [C-terminus of the Hsc (heat-shock cognate) 70-interacting protein] and c-Cbl (casitas B-lineage lymphoma). We demonstrate that CHIP and c-Cbl depletion have no effect on CFTR endocytosis, but c-Cbl depletion modestly enhanced the half-life of CFTR. The results of the present study define a significant role for Dab2 both in the endocytosis and post-endocytic fate of CFTR.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Endocitosis/fisiología , Complejo 2 de Proteína Adaptadora/metabolismo , Subunidades mu de Complejo de Proteína Adaptadora/metabolismo , Proteínas Reguladoras de la Apoptosis , Células Cultivadas , Regulador de Conductancia de Transmembrana de Fibrosis Quística/fisiología , Humanos , Transporte de Proteínas , Proteínas Proto-Oncogénicas c-cbl/metabolismo , ARN Interferente Pequeño/metabolismo , Proteínas Supresoras de Tumor , Ubiquitina-Proteína Ligasas/metabolismo
5.
J Mol Med (Berl) ; 101(9): 1141-1151, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37584738

RESUMEN

Autosomal-recessive polycystic kidney disease (ARPKD; MIM #263200) is a severe, hereditary, hepato-renal fibrocystic disorder that causes early childhood morbidity and mortality. Mutations in the polycystic kidney and hepatic disease 1 (PKHD1) gene, which encodes the protein fibrocystin/polyductin complex (FPC), cause all typical forms of ARPKD. Several mouse lines carrying diverse, genetically engineered disruptions in the orthologous Pkhd1 gene have been generated, but none expresses the classic ARPKD renal phenotype. In the current study, we characterized a spontaneous mouse Pkhd1 mutation that is transmitted as a recessive trait and causes cysticliver (cyli), similar to the hepato-biliary disease in ARPKD, but which is exacerbated by age, sex, and parity. We mapped the mutation to Chromosome 1 and determined that an insertion/deletion mutation causes a frameshift within Pkhd1 exon 48, which is predicted to result in a premature termination codon (UGA). Pkhd1cyli/cyli (cyli) mice exhibit a severe liver pathology but lack renal disease. Further analysis revealed that several alternatively spliced Pkhd1 mRNA, all containing exon 48, were expressed in cyli kidneys, but in lower abundance than in wild-type kidneys, suggesting that these transcripts escaped from nonsense-mediated decay (NMD). We identified an AAAAAT motif in exon 48 upstream of the cyli mutation which could enable ribosomal frameshifting, thus potentially allowing production of sufficient amounts of FPC for renoprotection. This mechanism, expressed in a species-specific fashion, may help explain the disparities in the renal phenotype observed between Pkhd1 mutant mice and patients with PKHD1-related disease. KEY MESSAGES: The Pkhd1cyli/cyli mouse expresses cystic liver disease, but no kidney phenotype. Pkhd1 mRNA expression is decreased in cyli liver and kidneys compared to wild-type. Ribosomal frameshifting may be responsible for Pkhd1 mRNA escape from NMD. Pkhd1 mRNA escape from NMD could contribute to the absent kidney phenotype.


Asunto(s)
Hepatopatías , Riñón Poliquístico Autosómico Recesivo , Preescolar , Ratones , Humanos , Animales , Riñón Poliquístico Autosómico Recesivo/genética , Riñón Poliquístico Autosómico Recesivo/patología , Riñón/metabolismo , Mutación , Factores de Transcripción/genética , ARN Mensajero/genética , Receptores de Superficie Celular/genética
6.
J Biol Chem ; 286(48): 41862-41870, 2011 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-22002058

RESUMEN

To identify endoplasmic reticulum (ER) stress-induced microRNAs (miRNA) that govern ER protein influx during the adaptive phase of unfolded protein response, we performed miRNA microarray profiling and analysis in human airway epithelial cells following ER stress induction using proteasome inhibition or tunicamycin treatment. We identified miR-346 as the most significantly induced miRNA by both classic stressors. miR-346 is encoded within an intron of the glutamate receptor ionotropic delta-1 gene (GRID1), but its ER stress-associated expression is independent of GRID1. We demonstrated that the spliced X-box-binding protein-1 (sXBP1) is necessary and sufficient for ER stress-associated miR-346 induction, revealing a novel role for this unfolded protein response-activated transcription factor. In mRNA profiling arrays, we identified 21 mRNAs that were reduced by both ER stress and miR-346. The target genes of miR-346 regulate immune responses and include the major histocompatibility complex (MHC) class I gene products, interferon-induced genes, and the ER antigen peptide transporter 1 (TAP1). Although most of the repressed mRNAs appear to be indirect targets because they lack specific seeding sites for miR-346, we demonstrate that the human TAP1 mRNA is a direct target of miR-346. The human TAP1 mRNA 3'-UTR contains a 6-mer canonical seeding site for miR-346. Importantly, the ER stress-associated reduction in human TAP1 mRNA and protein levels could be reversed with an miR-346 antagomir. Because TAP function is necessary for proper MHC class I-associated antigen presentation, our results provide a novel mechanistic explanation for reduced MHC class I-associated antigen presentation that was observed during ER stress.


Asunto(s)
Regiones no Traducidas 3'/fisiología , Transportadoras de Casetes de Unión a ATP/biosíntesis , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica/fisiología , MicroARNs/metabolismo , Factores de Transcripción/metabolismo , Respuesta de Proteína Desplegada/fisiología , Transportador de Casetes de Unión a ATP, Subfamilia B, Miembro 2 , Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/inmunología , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/inmunología , Perfilación de la Expresión Génica , Células HeLa , Antígenos de Histocompatibilidad Clase I/biosíntesis , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , MicroARNs/genética , MicroARNs/inmunología , Factores de Transcripción del Factor Regulador X , Factores de Transcripción/genética , Factores de Transcripción/inmunología , Proteína 1 de Unión a la X-Box
7.
Am J Physiol Lung Cell Mol Physiol ; 302(11): L1141-6, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22492740

RESUMEN

Cystic fibrosis (CF) is caused by the loss of the cystic fibrosis transmembrane conductance regulator (CFTR) function and results in a respiratory phenotype that is characterized by dehydrated mucus and bacterial infections that affect CF patients throughout their lives. Much of the morbidity and mortality in CF results from a failure to clear bacteria from the lungs. What causes the defect in the bacterial clearance in the CF lung has been the subject of an ongoing debate. Here we discuss the arguments for and against the role of the epithelial sodium channel, ENaC, in the development of CF lung disease.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Fibrosis Quística/metabolismo , Canales Epiteliales de Sodio/metabolismo , Animales , Bacterias/inmunología , Bacterias/patogenicidad , Infecciones Bacterianas/metabolismo , Infecciones Bacterianas/patología , Fibrosis Quística/genética , Fibrosis Quística/patología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/deficiencia , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Modelos Animales de Enfermedad , Canales Epiteliales de Sodio/genética , Humanos , Transporte Iónico , Pulmón/metabolismo , Pulmón/patología , Enfermedades Pulmonares/genética , Enfermedades Pulmonares/metabolismo , Enfermedades Pulmonares/patología , Ratones , Ratones Transgénicos , Porcinos
8.
J Biol Chem ; 285(37): 28741-8, 2010 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-20628052

RESUMEN

Recent advances in our understanding of translational dynamics indicate that codon usage and mRNA secondary structure influence translation and protein folding. The most frequent cause of cystic fibrosis (CF) is the deletion of three nucleotides (CTT) from the cystic fibrosis transmembrane conductance regulator (CFTR) gene that includes the last cytosine (C) of isoleucine 507 (Ile507ATC) and the two thymidines (T) of phenylalanine 508 (Phe508TTT) codons. The consequences of the deletion are the loss of phenylalanine at the 508 position of the CFTR protein (DeltaF508), a synonymous codon change for isoleucine 507 (Ile507ATT), and protein misfolding. Here we demonstrate that the DeltaF508 mutation alters the secondary structure of the CFTR mRNA. Molecular modeling predicts and RNase assays support the presence of two enlarged single stranded loops in the DeltaF508 CFTR mRNA in the vicinity of the mutation. The consequence of DeltaF508 CFTR mRNA "misfolding" is decreased translational rate. A synonymous single nucleotide variant of the DeltaF508 CFTR (Ile507ATC), that could exist naturally if Phe-508 was encoded by TTC, has wild type-like mRNA structure, and enhanced expression levels when compared with native DeltaF508 CFTR. Because CFTR folding is predominantly cotranslational, changes in translational dynamics may promote DeltaF508 CFTR misfolding. Therefore, we propose that mRNA "misfolding" contributes to DeltaF508 CFTR protein misfolding and consequently to the severity of the human DeltaF508 phenotype. Our studies suggest that in addition to modifier genes, SNPs may also contribute to the differences observed in the symptoms of various DeltaF508 homozygous CF patients.


Asunto(s)
Secuencia de Bases , Regulación de la Expresión Génica , Modelos Moleculares , Conformación de Ácido Nucleico , Polimorfismo de Nucleótido Simple , Pliegue de Proteína , ARN Mensajero/metabolismo , Eliminación de Secuencia , Células HeLa , Humanos , ARN Mensajero/genética
9.
Am J Respir Cell Mol Biol ; 42(3): 363-72, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19502384

RESUMEN

The most common mutation in the cystic fibrosis (CF) transmembrane conductance regulator (CFTR) gene, Delta F508, results in the production of a misfolded protein that is rapidly degraded. The mutant protein is temperature sensitive, and prior studies indicate that the low-temperature-rescued channel is poorly responsive to physiological stimuli, and is rapidly degraded from the cell surface at 37 degrees C. In the present studies, we tested the effect of a recently characterized pharmacological corrector, 2-(5-chloro-2-methoxy-phenylamino)-4'-methyl-[4,5'bithiazolyl-2'-yl]-phenyl-methanone (corr-4a), on cell surface stability and function of the low-temperature-rescued Delta F508 CFTR. We demonstrate that corr-4a significantly enhanced the protein stability of rescued Delta F508 CFTR for up to 12 hours at 37 degrees C (P < 0.05). Using firefly luciferase-based reporters to investigate the mechanisms by which low temperature and corr-4a enhance rescue, we found that low-temperature treatment inhibited proteasomal function, whereas corr-4a treatment inhibited the E1-E3 ubiquitination pathway. Ussing chamber studies indicated that corr-4a increased the cAMP-mediated Delta F508 CFTR response by 61% at 6 hours (P < 0.05), but not at later time points. However, addition of the CFTR channel activator, 4-methyl-2-(5-phenyl-1H-pyrazol-3-yl)-phenol, significantly augmented cAMP-stimulated currents, revealing that the biochemically detectable cell surface Delta F508 CFTR could be stimulated under the right conditions. Our studies demonstrate that stabilizing rescued Delta F508 CFTR was not sufficient to obtain maximal Delta F508 CFTR function in airway epithelial cells. These results strongly support the idea that maximal correction of Delta F508 CFTR requires a chemical corrector that: (1) promotes folding and exit from the endoplasmic reticulum; (2) enhances surface stability; and (3) improves channel activity.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Células Epiteliales/metabolismo , Estabilidad Proteica , Mucosa Respiratoria/metabolismo , Línea Celular , Polaridad Celular/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Humanos , Fenoles/farmacología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Estabilidad Proteica/efectos de los fármacos , Mucosa Respiratoria/efectos de los fármacos , Mucosa Respiratoria/patología , Temperatura , Factores de Tiempo , Ubiquitina/metabolismo , Ubiquitinación/efectos de los fármacos
10.
Expert Rev Proteomics ; 7(4): 495-506, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20653506

RESUMEN

Proteomic analysis has proved to be an important tool for understanding the complex nature of genetic disorders, such as cystic fibrosis (CF), by defining the cellular protein environment (proteome) associated with wild-type and mutant proteins. Proteomic screens identified the proteome of CF transmembrane conductance regulator (CFTR), and provided fundamental information to studies designed for understanding the crucial components of physiological CFTR function. Simultaneously, high-throughput screens for small-molecular correctors of CFTR mutants provided promising candidates for therapy. The majority of CF cases are caused by nucleotide deletions (DeltaF508 CFTR; >75%), resulting in CFTR misfolding, or insertion of premature termination codons ( approximately 10%), leading to unstable mRNA and reduced levels of truncated dysfunctional CFTR. In this article, we review recent results of proteomic screens, developments in identifying correctors for the most frequent CFTR mutants, and comment on how integration of the knowledge gained from these studies may aid in finding a cure for CF and a number of other genetic disorders.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/efectos de los fármacos , Fibrosis Quística/tratamiento farmacológico , Proteómica/métodos , Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/fisiología , Descubrimiento de Drogas/métodos , Evaluación Preclínica de Medicamentos/métodos , Humanos , Mutación
11.
Biochem J ; 410(3): 555-64, 2008 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-18052931

RESUMEN

Misfolded proteins destined for the cell surface are recognized and degraded by the ERAD [ER (endoplasmic reticulum) associated degradation] pathway. TS (temperature-sensitive) mutants at the permissive temperature escape ERAD and reach the cell surface. In this present paper, we examined a TS mutant of the CFTR [CF (cystic fibrosis) transmembrane conductance regulator], CFTR DeltaF508, and analysed its cell-surface trafficking after rescue [rDeltaF508 (rescued DeltaF508) CFTR]. We show that rDeltaF508 CFTR endocytosis is 6-fold more rapid (approximately 30% per 2.5 min) than WT (wild-type, approximately 5% per 2.5 min) CFTR at 37 degrees C in polarized airway epithelial cells (CFBE41o-). We also investigated rDeltaF508 CFTR endocytosis under two further conditions: in culture at the permissive temperature (27 degrees C) and following treatment with pharmacological chaperones. At low temperature, rDeltaF508 CFTR endocytosis slowed to WT rates (20% per 10 min), indicating that the cell-surface trafficking defect of rDeltaF508 CFTR is TS. Furthermore, rDeltaF508 CFTR is stabilized at the lower temperature; its half-life increases from <2 h at 37 degrees C to >8 h at 27 degrees C. Pharmacological chaperone treatment at 37 degrees C corrected the rDeltaF508 CFTR internalization defect, slowing endocytosis from approximately 30% per 2.5 min to approximately 5% per 2.5 min, and doubled DeltaF508 surface half-life from 2 to 4 h. These effects are DeltaF508 CFTR-specific, as pharmacological chaperones did not affect WT CFTR or transferrin receptor internalization rates. The results indicate that small molecular correctors may reproduce the effect of incubation at the permissive temperature, not only by rescuing DeltaF508 CFTR from ERAD, but also by enhancing its cell-surface stability.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Animales , Western Blotting , Cricetinae , Regulador de Conductancia de Transmembrana de Fibrosis Quística/efectos de los fármacos , Endocitosis , Semivida , Células HeLa , Humanos , Inmunoprecipitación , Temperatura
12.
Hum Mutat ; 29(9): 1118-24, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18570184

RESUMEN

Two major types of genetic variation are known: single nucleotide polymorphisms (SNPs), and a more recently discovered structural variation, involving changes in copy number (CNVs) of kilobase- to megabase-sized chromosomal segments. It is unknown whether CNVs arise in somatic cells, but it is, however, generally assumed that normal cells are genetically identical. We tested 34 tissue samples from three subjects and, having analyzed for each tissue < or =10(-6) of all cells expected in an adult human, we observed at least six CNVs, affecting a single organ or one or more tissues of the same subject. The CNVs ranged from 82 to 176 kb, often encompassing known genes, potentially affecting gene function. Our results indicate that humans are commonly affected by somatic mosaicism for stochastic CNVs, which occur in a substantial fraction of cells. The majority of described CNVs were previously shown to be polymorphic between unrelated subjects, suggesting that some CNVs previously reported as germline might represent somatic events, since in most studies of this kind, only one tissue is typically examined and analysis of parents for the studied subjects is not routinely performed. A considerable number of human phenotypes are a consequence of a somatic process. Thus, our conclusions will be important for the delineation of genetic factors behind these phenotypes. Consequently, biobanks should consider sampling multiple tissues to better address mosaicism in the studies of somatic disorders.


Asunto(s)
Dosificación de Gen , Mosaicismo , Polimorfismo Genético , Adulto , Cromosomas Humanos , Predisposición Genética a la Enfermedad , Genómica , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos , Especificidad de Órganos , Distribución Tisular
13.
Am J Respir Cell Mol Biol ; 39(4): 448-57, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18458236

RESUMEN

Environmental insults and misfolded proteins cause endoplasmic reticulum (ER) stress and activate the unfolded protein response (UPR). The UPR decreases endogenous cystic fibrosis transmembrane conductance regulator (CFTR) mRNA levels and protein maturation efficiency. Herein, we investigated the effects of the folding-deficient deltaF508 CFTR on ER stress induction and UPR activation. For these studies, we developed and characterized stable clones of Calu3deltaF cells that express different levels of endogenous wild-type (WT) and recombinant deltaF508 CFTR. We also present a novel RT-PCR-based assay for differential quantification of wild-type CFTR mRNA in the presence of deltaF508 CFTR message. The assay is based on a TaqMan minor groove binding (MGB) probe that recognizes a specific TTT sequence (encoding phenylalanine at position 508 in human CFTR). The MGB probe is extremely specific and sensitive to changes in WT CFTR message levels. In RNA samples that contain both WT and deltaF508 CFTR mRNAs, measurement of WT CFTR mRNA levels (using the MGB probe) and total CFTR mRNA (using commercial primers) allowed us to calculate deltaF508 CFTR mRNA levels. The results indicate that overexpression of deltaF508 CFTR causes ER stress and activates the UPR. UPR activation precedes a marked decrease in endogenous WT CFTR mRNA expression. Furthermore, polarized airway epithelial cell lines are important tools in cystic fibrosis research, and herein we provide an airway epithelial model to study the biogenesis and function of WT and deltaF508 CFTR expressed within the same cell.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/fisiología , Pliegue de Proteína , ARN Mensajero/metabolismo , Línea Celular , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Retículo Endoplásmico/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Humanos , Mutación , Mucosa Respiratoria/citología , Transducción de Señal
14.
N Engl J Med ; 349(15): 1433-41, 2003 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-14534336

RESUMEN

BACKGROUND: Mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene containing a premature termination signal cause a deficiency or absence of functional chloride-channel activity. Aminoglycoside antibiotics can suppress premature termination codons, thus permitting translation to continue to the normal end of the transcript. We assessed whether topical administration of gentamicin to the nasal epithelium of patients with cystic fibrosis could result in the expression of functional CFTR channels. METHODS: In a double-blind, placebo-controlled, crossover trial, patients with stop mutations in CFTR or patients homozygous for the DeltaF508 mutation received two drops containing gentamicin (0.3 percent, or 3 mg per milliliter) or placebo in each nostril three times daily for two consecutive periods of 14 days. Nasal potential difference was measured at base line and after each treatment period. Nasal epithelial cells were obtained before and after gentamicin treatment from patients carrying stop mutations, and the C-terminal of surface CFTR was stained. RESULTS: Gentamicin treatment caused a significant reduction in basal potential difference in the 19 patients carrying stop mutations (from -45+/-8 to -34+/-11 mV, P=0.005) and a significant response to chloride-free isoproterenol solution (from 0+/-3.6 to -5+/-2.7 mV, P<0.001). This effect of gentamicin on nasal potential difference occurred both in patients who were homozygous for stop mutations and in those who were heterozygous, but not in patients who were homozygous for DeltaF508. After gentamicin treatment, a significant increase in peripheral and surface staining for CFTR was observed in the nasal epithelial cells of patients carrying stop mutations. CONCLUSIONS: In patients with cystic fibrosis who have premature stop codons, gentamicin can cause translational "read through," resulting in the expression of full-length CFTR protein at the apical cell membrane, and thus can correct the typical electrophysiological abnormalities caused by CFTR dysfunction.


Asunto(s)
Antibacterianos/uso terapéutico , Codón sin Sentido/efectos de los fármacos , Regulador de Conductancia de Transmembrana de Fibrosis Quística/efectos de los fármacos , Fibrosis Quística/tratamiento farmacológico , Gentamicinas/uso terapéutico , Administración Intranasal , Adolescente , Adulto , Antibacterianos/farmacología , Niño , Estudios Cruzados , Fibrosis Quística/genética , Fibrosis Quística/fisiopatología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Eliminación de Gen , Genotipo , Gentamicinas/farmacología , Humanos , Potenciales de la Membrana/efectos de los fármacos , Persona de Mediana Edad , Mucosa Nasal/citología , Mucosa Nasal/efectos de los fármacos , Mucosa Nasal/fisiopatología
15.
J Cyst Fibros ; 6(5): 351-9, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17329172

RESUMEN

Quinine has been increasingly utilized as a placebo in cystic fibrosis (CF) clinical trials, including those leading to FDA approval of inhaled tobramycin, recent studies of anti-inflammatory aerosols such as glutathione, and clinical testing of hypertonic saline aerosols to augment mucous clearance. The drug effectively masks taste of experimental therapeutics, but could also confer changes in processes contributing to CF pathogenesis, including chloride secretion and paracellular ion permeability. In the Ussing chamber, concentrations of quinine (1 mg/ml) anticipated in the airways of CF subjects after aerosolization led to changes in chloride transport in Calu-3 (airway serous glandular) cell monolayers. Tissue resistance was significantly disrupted by the compound in both Calu-3 and primary airway epithelial cells in vitro. Lower doses of quinine (between 10 and 100 microg/ml) strongly inhibited the chloride secretory mechanism that utilizes CFTR, and forskolin activated I(SC) was reduced by approximately 24% and 44% in the presence of 10 and 100 microg/ml quinine, respectively. Our findings indicate that quinine disrupts airway epithelial functional integrity and blocks transepithelial chloride transport. The use of quinine as a taste-masking agent may have bioelectric effects relevant to CF trials using aerosolized drug delivery.


Asunto(s)
Cloruros/metabolismo , Fibrosis Quística/metabolismo , Células Epiteliales/efectos de los fármacos , Quinina/farmacología , Sistema Respiratorio/efectos de los fármacos , Células Cultivadas , Regulador de Conductancia de Transmembrana de Fibrosis Quística/fisiología , Humanos , Permeabilidad/efectos de los fármacos , Placebos
16.
Cancer Res ; 64(18): 6610-5, 2004 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-15374975

RESUMEN

Escherichia coli purine nucleoside phosphorylase (PNP) expressed in tumors converts relatively nontoxic prodrugs into membrane-permeant cytotoxic compounds with high bystander activity. In the present study, we examined tumor regressions resulting from treatment with E. coli PNP and fludarabine phosphate (F-araAMP), a clinically approved compound used in the treatment of hematologic malignancies. We tested bystander killing with an adenoviral construct expressing E. coli PNP and then more formally examined thresholds for the bystander effect, using both MuLv and lentiviral vectoring. Because of the importance of understanding the mechanism of bystander action and the limits to this anticancer strategy, we also evaluated in vivo variables related to the expression of E. coli PNP (level of E. coli PNP activity in tumors, ectopic expression in liver, percentage of tumor cells transduced in situ, and accumulation of active metabolites in tumors). Our results indicate that F-araAMP confers excellent in vivo dose-dependent inhibition of bystander tumor cells, including strong responses in subcutaneous human glioma xenografts when 95 to 97.5% of the tumor mass is composed of bystander cells. These findings define levels of E. coli PNP expression necessary for antitumor activity with F-araAMP and demonstrate new potential for a clinically approved compound in solid tumor therapy.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Escherichia coli/genética , Terapia Genética/métodos , Purina-Nucleósido Fosforilasa/genética , Fosfato de Vidarabina/análogos & derivados , Fosfato de Vidarabina/farmacología , Adenoviridae/genética , Animales , Antimetabolitos Antineoplásicos/farmacocinética , Línea Celular Tumoral , Terapia Combinada , Relación Dosis-Respuesta a Droga , Escherichia coli/enzimología , Vectores Genéticos/genética , Glioma/tratamiento farmacológico , Glioma/enzimología , Glioma/genética , Humanos , Lentivirus/genética , Ratones , Ratones Desnudos , Virus de la Leucemia Murina de Moloney/genética , Purina-Nucleósido Fosforilasa/biosíntesis , Purina-Nucleósido Fosforilasa/metabolismo , Transfección/métodos , Fosfato de Vidarabina/farmacocinética , Ensayos Antitumor por Modelo de Xenoinjerto
17.
J Mol Med (Berl) ; 80(9): 595-604, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12226741

RESUMEN

Cystic fibrosis (CF) is caused by mutations in the gene encoding the cystic fibrosis transmembrane conductance regulator (CFTR) protein. Since approximately 5% of all mutant CF alleles are stop mutations, it can be calculated that approximately 10% of CF patients carry a premature stop mutation in at least one copy of the CFTR gene. Certain ethnic groups, such as the Ashkenazi Jewish population, carry a much higher percentage of CF stop mutations. Consequently, a therapeutic strategy aimed at suppressing this class of mutation would be highly desirable for the treatment of this common genetic disease. We have shown previously that aminoglycoside antibiotics can suppress premature stop mutations in the CFTR gene in a bronchial epithelial cell line [Nat Med (1997) 3:1280]. To address whether aminoglycosides can suppress a CFTR premature stop mutation in an animal model, we constructed a transgenic mouse with a null mutation in the endogenous CFTR locus (Cftr-/-) that also expressed a human CFTR-G542X cDNA under control of the intestinal fatty acid binding protein promoter. We then investigated whether the daily administration of the aminoglycoside antibiotics gentamicin or tobramycin could restore the expression of a detectable level of CFTR protein. Immunofluorescence staining of intestinal tissues from Cftr-/- hCFTR-G542X mice revealed that gentamicin treatment resulted in the appearance of hCFTR protein at the apical surface of the glands of treated mice. Weaker staining was also observed in the intestinal glands following tobramycin treatment. Short-circuit current measurements made on intestinal tissues from these mice demonstrated that a significant number of positive cAMP-stimulated transepithelial chloride current measurements could be observed following gentamicin treatment (P=0.008) and a near significant number following tobramycin treatment (P=0.052). When taken together, these results indicate that gentamicin, and to a lesser extent tobramycin, can restore the synthesis of functional hCFTR protein by suppressing the hCFTR-G542X premature stop mutation in vivo.


Asunto(s)
Antibacterianos/farmacología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Fibrosis Quística/genética , Mutación , Supresión Genética , Transgenes , Potenciales de Acción/efectos de los fármacos , Animales , Colforsina/farmacología , Fibrosis Quística/patología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Expresión Génica , Gentamicinas/administración & dosificación , Gentamicinas/sangre , Gentamicinas/farmacología , Heterocigoto , Humanos , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Ratones , Ratones Noqueados , Ratones Transgénicos , Regiones Promotoras Genéticas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Tobramicina/administración & dosificación , Tobramicina/farmacología
18.
PLoS One ; 10(4): e0123131, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25879443

RESUMEN

The ΔF508 mutant form of the cystic fibrosis transmembrane conductance regulator (ΔF508 CFTR) that is normally degraded by the ER-associated degradative pathway can be rescued to the cell surface through low-temperature (27°C) culture or small molecular corrector treatment. However, it is unstable on the cell surface, and rapidly internalized and targeted to the lysosomal compartment for degradation. To understand the mechanism of this rapid turnover, we examined the role of two adaptor complexes (AP-2 and Dab2) and three E3 ubiquitin ligases (c-Cbl, CHIP, and Nedd4-2) on low-temperature rescued ΔF508 CFTR endocytosis and degradation in human airway epithelial cells. Our results demonstrate that siRNA depletion of either AP-2 or Dab2 inhibits ΔF508 CFTR endocytosis by 69% and 83%, respectively. AP-2 or Dab2 depletion also increases the rescued protein half-life of ΔF508 CFTR by ~18% and ~91%, respectively. In contrast, the depletion of each of the E3 ligases had no effect on ΔF508 CFTR endocytosis, whereas CHIP depletion significantly increased the surface half-life of ΔF508 CFTR. To determine where and when the ubiquitination occurs during ΔF508 CFTR turnover, we monitored the ubiquitination of rescued ΔF508 CFTR during the time course of CFTR endocytosis. Our results indicate that ubiquitination of the surface pool of ΔF508 CFTR begins to increase 15 min after internalization, suggesting that CFTR is ubiquitinated in a post-endocytic compartment. This post-endocytic ubiquination of ΔF508 CFTR could be blocked by either inhibiting endocytosis, by siRNA knockdown of CHIP, or by treating cells with the CFTR corrector, VX-809. Our results indicate that the post-endocytic ubiquitination of CFTR by CHIP is a critical step in the peripheral quality control of cell surface ΔF508 CFTR.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Proteínas de Drosophila/fisiología , Endocitosis , Proteínas Nucleares/fisiología , Proteínas Supresoras de Tumor/fisiología , Proteínas Reguladoras de la Apoptosis , Células Cultivadas , Regulador de Conductancia de Transmembrana de Fibrosis Quística/química , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Humanos , ARN Interferente Pequeño/genética , Propiedades de Superficie , Ubiquitinación
20.
Methods Enzymol ; 491: 3-24, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21329791

RESUMEN

The cystic fibrosis transmembrane conductance regulator (CFTR) is a chloride channel and key regulator of epithelial functions. Mutations in the CFTR gene lead to reduced or dysfunctional CFTR protein and cause cystic fibrosis (CF), a generalized exocrinopathy affecting multiple organs. In the airways, loss of CFTR function leads to thickened mucus, reduced mucociliary clearance, chronic infections, and respiratory failure. Common airway disorders such as bronchitis and chronic obstructive pulmonary disease (COPD) also present CF-like symptoms such as mucus congestion and chronic inflammation without mutations in CFTR. The primary risk factors for COPD and chronic bronchitis include environmental stress insults such as pollutants and infections that often result in hypoxic conditions. Furthermore, environmental factors such as cigarette smoke and reactive oxygen species have been implicated in reduced CFTR function. Activation of cellular stress responses by these factors promotes differential, stress-associated gene expression regulation. During our investigations on the mechanisms of CFTR expression regulation, we have shown that the ER stress response, the unfolded protein response (UPR), decreases CFTR expression at the transcriptional, translational, and maturational levels. Here, we provide a detailed description of the methods we employ to study CFTR expression regulation by the UPR. Similar approaches are applicable in studies on other genes and how they are affected by the UPR.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Retículo Endoplásmico/genética , Técnicas Genéticas , Respuesta de Proteína Desplegada , Animales , Western Blotting/métodos , Línea Celular , Inmunoprecipitación de Cromatina/métodos , Fibrosis Quística/genética , Fibrosis Quística/metabolismo , Retículo Endoplásmico/metabolismo , Células Epiteliales/metabolismo , Humanos , ARN Mensajero/genética , ARN Mensajero/aislamiento & purificación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA