Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 182
Filtrar
1.
J Lipid Res ; 65(7): 100551, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39002195

RESUMEN

Intestinal disease is one of the earliest manifestations of cystic fibrosis (CF) in children and is closely tied to deficits in growth and nutrition, both of which are directly linked to future mortality. Patients are treated aggressively with pancreatic enzyme replacement therapy and a high-fat diet to circumvent fat malabsorption, but this does not reverse growth and nutritional defects. We hypothesized that defects in chylomicron production could explain why CF body weights and nutrition are so resistant to clinical treatments. We used gold standard intestinal lipid absorption and metabolism approaches, including mouse mesenteric lymph cannulation, in vivo chylomicron secretion kinetics, transmission electron microscopy, small intestinal organoids, and chylomicron metabolism assays to test this hypothesis. In mice expressing the G542X mutation in cystic fibrosis transmembrane conductance regulator (CFTR-/- mice), we find that defective FFA trafficking across the epithelium into enterocytes drives a chylomicron formation defect. Furthermore, G542X mice secrete small, triglyceride-poor chylomicrons into the lymph and blood. These defective chylomicrons are cleared into extraintestinal tissues at ∼10-fold faster than WT chylomicrons. This defect in FFA absorption resulting in dysfunctional chylomicrons cannot be explained by steatorrhea or pancreatic insufficiency and is maintained in primary small intestinal organoids treated with micellar lipids. These studies suggest that the ultrahigh-fat diet that most people with CF are counselled to follow may instead make steatorrhea and malabsorption defects worse by overloading the absorptive capacity of the CF small intestine.


Asunto(s)
Quilomicrones , Fibrosis Quística , Fibrosis Quística/metabolismo , Fibrosis Quística/patología , Fibrosis Quística/genética , Animales , Quilomicrones/metabolismo , Ratones , Ácidos Grasos no Esterificados/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/deficiencia , Insuficiencia Pancreática Exocrina/metabolismo , Insuficiencia Pancreática Exocrina/genética , Insuficiencia Pancreática Exocrina/patología , Transporte Biológico , Humanos , Mucosa Intestinal/metabolismo
2.
Eur J Pharmacol ; 978: 176771, 2024 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-38925289

RESUMEN

The CFTR modulator Trikafta has markedly improved lung disease for Cystic Fibrosis (CF) patients carrying the common delta F508 (F508del-CFTR) CFTR mutation. F508del-CFTR results in an apical trafficking defect and loss of function in CFTR-expressing epithelial cells. However, Trikafta has not resulted in improved gastrointestinal function in CF patients. A humanized mouse model of F508del-CFTR was recently generated to evaluate CFTR modulators and other compounds to treat human F508del-CFTR CF intestinal disease. Short-term (4 h) treatment of rats with Dexamethasone (Dex) potently activates serum glucocorticoid kinase 1 (SGK1) and increases CFTR apical traffic and ion transport in the native intestine. This study examined CFTR localization and ion transport in intestinal segments from humanized F508del-CFTR mice following treatment with Dex in the presence/absence of Trikafta. Dex treatment improved apical CFTR localization and function but was inconsistent along intestinal segments. Combined treatment with Dex and Trikafta was superior to Dex alone but inconsistently improved CFTR localization and function. These data suggest further optimization of humanized CF mouse models will be necessary to test the efficacy of compounds to treat human CF intestinal disease.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística , Dexametasona , Animales , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/deficiencia , Humanos , Ratones , Dexametasona/farmacología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/deficiencia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/efectos de los fármacos , Intestinos/efectos de los fármacos , Fibrosis Quística/tratamiento farmacológico , Fibrosis Quística/metabolismo , Fibrosis Quística/genética , Activación Enzimática/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Mutación , Masculino , Indoles , Benzodioxoles
3.
Cell Rep ; 37(1): 109795, 2021 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-34610318

RESUMEN

A controversial hypothesis pertaining to cystic fibrosis (CF) lung disease is that the CF transmembrane conductance regulator (CFTR) channel fails to inhibit the epithelial Na+ channel (ENaC), yielding increased Na+ reabsorption and airway dehydration. We use a non-invasive self-referencing Na+-selective microelectrode technique to measure Na+ transport across individual folds of distal airway surface epithelium preparations from CFTR-/- (CF) and wild-type (WT) swine. We show that, under unstimulated control conditions, WT and CF epithelia exhibit similar, low rates of Na+ transport that are unaffected by the ENaC blocker amiloride. However, in the presence of the cyclic AMP (cAMP)-elevating agents forskolin+IBMX (isobutylmethylxanthine), folds of WT tissues secrete large amounts of Na+, while CFTR-/- tissues absorb small, but potentially important, amounts of Na+. In cAMP-stimulated conditions, amiloride inhibits Na+ absorption in CFTR-/- tissues but does not affect secretion in WT tissues. Our results are consistent with the hypothesis that ENaC-mediated Na+ absorption may contribute to dehydration of CF distal airways.


Asunto(s)
AMP Cíclico/metabolismo , Canales Epiteliales de Sodio/metabolismo , Epitelio/metabolismo , Sodio/metabolismo , 1-Metil-3-Isobutilxantina/farmacología , Amilorida/farmacología , Animales , Animales Modificados Genéticamente/metabolismo , Colforsina/farmacología , Fibrosis Quística , Regulador de Conductancia de Transmembrana de Fibrosis Quística/deficiencia , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Modelos Animales de Enfermedad , Bloqueadores del Canal de Sodio Epitelial/farmacología , Canales Epiteliales de Sodio/química , Transporte Iónico/efectos de los fármacos , Masculino , Porcinos
4.
Physiol Rep ; 9(14): e14978, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34288572

RESUMEN

Hepatobiliary disease causes significant morbidity and mortality in people with cystic fibrosis (CF), yet this problem remains understudied. Previous studies in the newborn CF pig demonstrated decreased bile flow into the small intestine and a microgallbladder with increased luminal mucus and fluid secretion defects. In this study, we examined the intrahepatic bile ducts of the newborn CF pig. We assessed whether our findings from the gallbladder are present elsewhere in the porcine biliary tract and if CF pig cholangiocytes have fluid secretion defects. Immunohistochemistry demonstrated apical CFTR expression in non-CF pig intrahepatic bile ducts of a variety of sizes; CF pig intrahepatic bile ducts lacked CFTR expression. Assessment of serum markers did not reveal significant signs of hepatobiliary disease except for an elevation in direct bilirubin. Quantitative histology demonstrated that CF pigs had smaller bile ducts that more frequently contained luminal mucus. CF intrahepatic cholangiocyte organoids were smaller and lacked cAMP-mediated fluid secretion. Together these data suggest that cholangiocyte fluid secretion is decreased in the CF pig, contributing to structural changes in bile ducts and decreased biliary flow.


Asunto(s)
Conductos Biliares Intrahepáticos/metabolismo , Conductos Biliares Intrahepáticos/patología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/deficiencia , Fibrosis Quística/metabolismo , Fibrosis Quística/patología , Modelos Animales de Enfermedad , Animales , Animales Modificados Genéticamente , Animales Recién Nacidos , Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Porcinos
5.
Am J Respir Cell Mol Biol ; 64(5): 604-616, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33616476

RESUMEN

Premature-termination codons (PTCs) in CFTR (cystic fibrosis [CF] transmembrane conductance regulator) result in nonfunctional CFTR protein and are the proximate cause of ∼11% of CF-causing alleles, for which no treatments exist. The CFTR corrector lumacaftor and the potentiator ivacaftor improve CFTR function with terminal PTC mutations and enhance the effect of readthrough agents. Novel correctors GLPG2222 (corrector 1 [C1]), GLPG3221 (corrector 2 [C2]), and potentiator GLPG1837 compare favorably with lumacaftor and ivacaftor in vitro. Here, we evaluated the effect of correctors C1a and C2a (derivatives of C1 and C2) and GLPG1837 alone or in combination with the readthrough compound G418 on CFTR function using heterologous Fischer rat thyroid (FRT) cells, the genetically engineered human bronchial epithelial (HBE) 16HBE14o- cell lines, and primary human cells with PTC mutations. In FRT lines pretreated with G418, GLPG1837 elicited dose-dependent increases in CFTR activity that exceeded those from ivacaftor in FRT-W1282X and FRT-R1162X cells. A three-mechanism strategy consisting of G418, GLPG1837, and two correctors (C1a + C2a) yielded the greatest functional improvements in FRT and 16HBE14o- PTC variants, noting that correction and potentiation without readthrough was sufficient to stimulate CFTR activity for W1282X cells. GLPG1837 + C1a + C2a restored substantial function in G542X/F508del HBE cells and restored even more function for W1282X/F508del cells, largely because of the corrector/potentiator effect, with no additional benefit from G418. In G542X/R553X or R1162X/R1162X organoids, enhanced forskolin-induced swelling was observed with G418 + GLPG1837 + C1a + C2a, although GLPG1837 + C1a + C2a alone was sufficient to improve forskolin-induced swelling in W1282X/W1282X organoids. Combination of CFTR correctors, potentiators, and readthrough compounds augments the functional repair of CFTR nonsense mutations, indicating the potential for novel correctors and potentiators to restore function to truncated W1282X CFTR.


Asunto(s)
Benzoatos/farmacología , Benzopiranos/farmacología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Fibrosis Quística/genética , Células Epiteliales/efectos de los fármacos , Biosíntesis de Proteínas/efectos de los fármacos , Piranos/farmacología , Pirazoles/farmacología , Aminofenoles/farmacología , Aminopiridinas/farmacología , Animales , Benzodioxoles/farmacología , Línea Celular , Cloruros/metabolismo , Codón sin Sentido , Fibrosis Quística/tratamiento farmacológico , Fibrosis Quística/metabolismo , Fibrosis Quística/patología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/agonistas , Regulador de Conductancia de Transmembrana de Fibrosis Quística/deficiencia , Células Epiteliales/metabolismo , Humanos , Transporte Iónico/efectos de los fármacos , Quinolonas/farmacología , Ratas , Recuperación de la Función , Células Epiteliales Tiroideas/efectos de los fármacos , Células Epiteliales Tiroideas/metabolismo
6.
J Cyst Fibros ; 20(1): 173-182, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32978064

RESUMEN

BACKGROUND: Bacterial colonization in cystic fibrosis (CF) lungs has been directly associated to the loss of CFTR function, and/or secondarily linked to repetitive cycles of chronic inflammation/infection. We hypothesized that altered molecular properties of mucins could contribute to this process. METHODS: Newborn CFTR+/+ and CFTR-/- were sacrificed before and 6 h after inoculation with luminescent Pseudomonas aeruginosa into the tracheal carina. Tracheal mucosa and the bronchoalveolar lavage (BAL) fluid were collected to determine the level of mucin O-glycosylation, bacteria binding to mucins and the airways transcriptome. Disturbances in mucociliary transport were determined by ex-vivo imaging of luminescent Pseudomonas aeruginosa. RESULTS: We provide evidence of an increased sialylation of CF airway mucins and impaired mucociliary transport that occur before the onset of inflammation. Hypersialylation of mucins was reproduced on tracheal explants from non CF animals treated with GlyH101, an inhibitor of CFTR channel activity, indicating a causal relationship between the absence of CFTR expression and the sialylation of mucins. This increased sialylation was correlated to an increased adherence of P. aeruginosa to mucins. In vivo infection of newborn CF piglets by live luminescent P. aeruginosa demonstrated an impairment of mucociliary transport of this bacterium, with no evidence of pre-existing inflammation. CONCLUSIONS: Our results document for the first time in a well-defined CF animal model modifications that affect the O-glycan chains of mucins. These alterations precede infection and inflammation of airway tissues, and provide a favorable context for microbial development in CF lung that hallmarks this disease.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/deficiencia , Fibrosis Quística/metabolismo , Fibrosis Quística/fisiopatología , Mucinas/metabolismo , Depuración Mucociliar , Mucosa Respiratoria/metabolismo , Animales , Animales Recién Nacidos , Femenino , Glicosilación , Masculino , Pseudomonas aeruginosa , Mucosa Respiratoria/microbiología , Porcinos , Tráquea
7.
Mol Vis ; 26: 780-788, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33311973

RESUMEN

Purpose: Vasoactive intestinal peptide (VIP) is an important regulator of lacrimal gland (LG) function although the effect of VIP on ductal fluid secretion is unknown. Therefore, the aim of the present study was to investigate the role of VIP in the regulation of fluid secretion of isolated LG ducts and to analyze the underlying intracellular mechanisms. Methods: LGs from wild-type (WT) and cystic fibrosis transmembrane conductance regulator (CFTR) knockout (KO) mice were used. Immunofluorescence was applied to confirm the presence of VIP receptors termed VPAC1 and VPAC2 in LG duct cells. Ductal fluid secretion evoked by VIP (100 nM) was measured in isolated ducts using videomicroscopy. Intracellular Ca2+ signaling underlying VIP stimulation was investigated with microfluorometry. Results: VIP stimulation resulted in a robust and continuous fluid secretory response in isolated duct segments originated from WT mice. In contrast, CFTR KO ducts exhibited only a weak pulse-like secretion. A small but statistically significant increase was detected in the intracellular Ca2+ level [Ca2+]i during VIP stimulation in the WT and in CFTR KO ducts. VIP-evoked changes in [Ca2+]i did not differ considerably between the WT and CFTR KO ducts. Conclusions: These results suggest the importance of VIP in the regulation of ductal fluid secretion and the determining role of the adenylyl cyclase-cAMP-CFTR route in this process.


Asunto(s)
Aparato Lagrimal/metabolismo , Lágrimas/metabolismo , Péptido Intestinal Vasoactivo/metabolismo , Animales , Calcio/metabolismo , Señalización del Calcio , Carbacol/farmacología , Quelantes/farmacología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/deficiencia , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Ácido Egtácico/análogos & derivados , Ácido Egtácico/metabolismo , Espacio Intracelular/metabolismo , Ratones Noqueados , Receptores de Tipo II del Péptido Intestinal Vasoactivo/metabolismo , Receptores de Tipo I del Polipéptido Intestinal Vasoactivo/metabolismo
8.
Hum Gene Ther ; 31(17-18): 921-939, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32814451

RESUMEN

The lung has long been a target for gene therapy, yet efficient delivery and phenotypic disease correction has remained challenging. Although there have been significant advancements in gene therapies of other organs, including the development of several ex vivo therapies, in vivo therapeutics of the lung have been slower to transition to the clinic. Within the past few years, the field has witnessed an explosion in the development of new gene addition and gene editing strategies for the treatment of monogenic disorders. In this review, we will summarize current developments in gene therapy for cystic fibrosis, alpha-1 antitrypsin deficiency, and surfactant protein deficiencies. We will explore the different gene addition and gene editing strategies under investigation and review the challenges of delivery to the lung.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Fibrosis Quística/terapia , Dependovirus/genética , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Proteína A Asociada a Surfactante Pulmonar/genética , alfa 1-Antitripsina/genética , Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/deficiencia , Edición Génica , Vectores Genéticos/genética , Humanos , Proteína A Asociada a Surfactante Pulmonar/deficiencia
9.
J Leukoc Biol ; 108(6): 1777-1785, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32531843

RESUMEN

Persistent neutrophilic inflammation is a hallmark of cystic fibrosis (CF). However, the mechanisms underlying this outstanding pathology remain incompletely understood. Here, we report that CFTR in myeloid immune cells plays a pivotal role in control of neutrophilic inflammation. Myeloid CFTR-Knockout (Mye-Cftr-/-) mice and congenic wild-type (WT) mice were challenged peritoneally with zymosan particles at different doses, creating aseptic peritonitis with varied severity. A high-dose challenge resulted in significantly higher mortality in Mye-Cftr-/- mice, indicating an intrinsic defect in host control of inflammation in mice whose myeloid cells lack CF. The low-dose challenge demonstrated an impaired resolution of inflammation in Mye-Cftr-/- mice, reflected by a significant overproduction of proinflammatory cytokines, including neutrophil chemokines MIP-2 and KC, and sustained accumulation of neutrophils. Tracing neutrophil mobilization in vivo demonstrated that myeloid CF mice recruited significantly more neutrophils than did WT mice. Pulmonary challenge with zymosan elicited exuberant inflammation in the lung and recapitulated the findings from peritoneal challenge. To determine the major type of cell that was primarily responsible for the over-recruitment of neutrophils, we purified and cultured ex vivo zymosan-elicited peritoneal neutrophils and macrophages. The CF neutrophils produced significantly more MIP-2 than did the WT counterparts, and peripheral blood neutrophils isolated from myeloid CF mice also produced significantly more MIP-2 after zymosan stimulation in vitro. These data altogether suggest that CFTR dysfunction in myeloid immune cells, especially neutrophils, leads to hyperinflammation and excessive neutrophil mobilization in the absence of infection. Thus, dysregulated inflammation secondary to abnormal or absent CFTR in myeloid cells may underlie the clinically observed neutrophilic inflammation in CF.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/deficiencia , Fibrosis Quística/inmunología , Macrófagos Peritoneales/inmunología , Neutrófilos/inmunología , Animales , Fibrosis Quística/patología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/inmunología , Inflamación/inducido químicamente , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Mutación con Pérdida de Función , Macrófagos Peritoneales/patología , Ratones , Ratones Mutantes , Neutrófilos/patología , Zimosan/toxicidad
10.
Genes (Basel) ; 11(5)2020 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-32403302

RESUMEN

Cystic fibrosis (CF) is a genetic disease with mutational changes leading to profound dysbiosis, both pulmonary and intestinal, from a very young age. This dysbiosis plays an important role in clinical manifestations, particularly in the lungs, affected by chronic infection. The range of microbiological tools has recently been enriched by metagenomics based on next-generation sequencing (NGS). Currently applied essentially in a gene-targeted manner, metagenomics has enabled very exhaustive description of bacterial communities in the CF lung niche and, to a lesser extent, the fungi. Aided by progress in bioinformatics, this now makes it possible to envisage shotgun sequencing and opens the door to other areas of the microbial world, the virome, and the archaeome, for which almost everything remains to be described in cystic fibrosis. Paradoxically, applying NGS in microbiology has seen a rebirth of bacterial culture, but in an extended manner (culturomics), which has proved to be a perfectly complementary approach to NGS. Animal models have also proved indispensable for validating microbiome pathophysiological hypotheses. Description of pathological microbiomes and correlation with clinical status and therapeutics (antibiotic therapy, cystic fibrosis transmembrane conductance regulator (CFTR) modulators) revealed the richness of microbiome data, enabling description of predictive and follow-up biomarkers. Although monogenic, CF is a multifactorial disease, and both genotype and microbiome profiles are crucial interconnected factors in disease progression. Microbiome-genome interactions are thus important to decipher.


Asunto(s)
Fibrosis Quística/microbiología , Disbiosis/complicaciones , Microbiota , Aminofenoles/farmacología , Animales , Archaea/aislamiento & purificación , Bacterias/aislamiento & purificación , Biomarcadores , Fibrosis Quística/genética , Fibrosis Quística/patología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/deficiencia , Regulador de Conductancia de Transmembrana de Fibrosis Quística/efectos de los fármacos , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Hongos/aislamiento & purificación , Microbioma Gastrointestinal/efectos de los fármacos , Genotipo , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Pulmón/microbiología , Pulmón/patología , Pulmón/virología , Mamíferos , Metagenómica , Microbiota/efectos de los fármacos , Especificidad de Órganos , Pronóstico , Quinolonas/farmacología , Virus/aislamiento & purificación
11.
Am J Physiol Lung Cell Mol Physiol ; 318(5): L931-L942, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32130033

RESUMEN

The human airway is protected by an efficient innate defense mechanism that requires healthy secretion of airway surface liquid (ASL) to clear pathogens from the lungs. Most of the ASL in the upper airway is secreted by submucosal glands. In cystic fibrosis (CF), the function of airway submucosal glands is abnormal, and these abnormalities are attributed to anomalies in ion transport across the epithelia lining the different sections of the glands that function coordinately to produce the ASL. However, the ion transport properties of most of the anatomical regions of the gland have never been measured, and there is controversy regarding which segments express CFTR. This makes it difficult to determine the glandular abnormalities that may contribute to CF lung disease. Using a noninvasive, extracellular self-referencing ion-selective electrode technique, we characterized ion transport properties in all four segments of submucosal glands from wild-type and CFTR-/- swine. In wild-type airways, the serous acini, mucus tubules, and collecting ducts secrete Cl- and Na+ into the lumen in response to carbachol and forskolin stimulation. The ciliated duct also transports Cl- and Na+ but in the opposite direction, i.e., reabsorption from the ASL, which may contribute to lowering Na+ and Cl- activities in the secreted fluid. In CFTR-/- airways, the serous acini, collecting ducts, and ciliated ducts fail to transport ions after forskolin stimulation, resulting in the production of smaller volumes of ASL with normal Cl-, Na+, and K+ concentration.


Asunto(s)
Células Acinares/metabolismo , Cilios/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Fibrosis Quística/metabolismo , Pulmón/metabolismo , Células Acinares/efectos de los fármacos , Células Acinares/patología , Animales , Carbacol/farmacología , Cationes Monovalentes , Cloruros/metabolismo , Cilios/efectos de los fármacos , Cilios/patología , Colforsina/farmacología , Fibrosis Quística/genética , Fibrosis Quística/patología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/deficiencia , Modelos Animales de Enfermedad , Técnicas Electroquímicas , Electrodos , Eliminación de Gen , Expresión Génica , Humanos , Transporte Iónico , Pulmón/efectos de los fármacos , Pulmón/patología , Potasio/metabolismo , Sodio/metabolismo , Porcinos
12.
Am J Pathol ; 190(5): 977-993, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32084371

RESUMEN

Animal models of cystic fibrosis (CF) are essential for investigating disease mechanisms and trialing potential therapeutics. This study generated two CF rat models using clustered regularly interspaced short palindromic repeats/clustered regularly interspaced short palindromic repeats-associated protein 9 gene editing. One rat model carries the common human Phe508del (ΔF508) CF transmembrane conductance regulator (CFTR) mutation, whereas the second is a CFTR knockout model. Phenotype was characterized using a range of functional and histologic assessments, including nasal potential difference to measure electrophysiological function in the upper airways, RNAscope in situ hybridization and quantitative PCR to assess CFTR mRNA expression in the lungs, immunohistochemistry to localize CFTR protein in the airways, and histopathologic assessments in a range of tissues. Both rat models revealed a range of CF manifestations, including reduced survival, intestinal obstruction, bioelectric defects in the nasal epithelium, histopathologic changes in the trachea, large intestine, and pancreas, and abnormalities in the development of the male reproductive tract. The CF rat models presented herein will prove useful for longitudinal assessments of pathophysiology and therapeutics.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/deficiencia , Fibrosis Quística , Modelos Animales de Enfermedad , Edición Génica/métodos , Animales , Sistemas CRISPR-Cas , Fibrosis Quística/genética , Fibrosis Quística/patología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Humanos , Ratones Noqueados , Mutación , Fenotipo , Ratas , Ratas Sprague-Dawley
14.
Genomics ; 112(2): 1139-1150, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31251978

RESUMEN

Cystic fibrosis (CF) is caused by mutations in the gene encoding the CFTR anion channel. Loss of CFTR function in pancreatic, biliary and intestinal epithelia, severely affects gastrointestinal function. Transcriptome analysis indicated the activation of an innate and adaptive immune response in the distal small intestine of Cftr null mice. Inflammation was associated with differential regulation of numerous genes involved in the transport and metabolism of nutrients and, particularly, lipids, that are targeted by ligand-dependent nuclear receptors and/or HNF4α. Among the most strongly down-regulated genes are the FXR targets Fgf15 and Nr0b2, the PPARα target Pdk4, and the PXR target Ces2a, whereas expression of the CF modifier gene Slc6a14 was strongly increased. Most changes in gene expression were reversed by bacterial containment. Our data suggest that the gut microbiota has a pervasive effect on gene expression in CF mice, affecting enterocyte maturation, lipid metabolism, and nutrient absorption in CF.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Intestino Delgado/metabolismo , Transcriptoma , Sistemas de Transporte de Aminoácidos/genética , Sistemas de Transporte de Aminoácidos/metabolismo , Animales , Carboxilesterasa/genética , Carboxilesterasa/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/deficiencia , Regulación hacia Abajo , Femenino , Factores de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/metabolismo , Microbioma Gastrointestinal , Eliminación de Gen , Factor Nuclear 4 del Hepatocito/genética , Factor Nuclear 4 del Hepatocito/metabolismo , Inmunidad Innata , Intestino Delgado/inmunología , Intestino Delgado/microbiología , Masculino , Ratones , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora/genética , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora/metabolismo , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo
15.
Am J Respir Cell Mol Biol ; 61(6): 755-764, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31189070

RESUMEN

SLC6A14-mediated l-arginine transport has been shown to augment the residual anion channel activity of the major mutant, F508del-CFTR, in the murine gastrointestinal tract. It is not yet known if this transporter augments residual and pharmacological corrected F508del-CFTR in primary airway epithelia. We sought to determine the role of l-arginine uptake via SLC6A14 in modifying F508del-CFTR channel activity in airway cells from patients with cystic fibrosis (CF). Human bronchial epithelial (HBE) cells from lung explants of patients without CF (HBE) and those with CF (CF-HBE) were used for H3-flux, airway surface liquid, and Ussing chamber studies. We used α-methyltryptophan as a specific inhibitor for SLC6A14. CFBE41o-, a commonly used CF airway cell line, was employed for studying the mechanism of the functional interaction between SLC6A14 and F508del-CFTR. SLC6A14 is functionally expressed in CF-HBE cells. l-arginine uptake via SLC6A14 augmented F508del-CFTR function at baseline and after treatment with lumacaftor. SLC6A14-mediated l-arginine uptake also increased the airway surface liquid in CF-HBE cells. Using CFBE41o cells, we showed that the positive SLC6A14 effect was mainly dependent on the nitric oxide (NO) synthase activity, nitrogen oxides, including NO, and phosphorylation by protein kinase G. These finding were confirmed in CF-HBE, as inducible NO synthase inhibition abrogated the functional interaction between SLC6A14 and pharmacological corrected F508del-CFTR. In summary, SLC6A14-mediated l-arginine transport augments residual F508del-CFTR channel function via a noncanonical, NO pathway. This effect is enhanced with increasing pharmacological rescue of F508del-CFTR to the membrane. The current study demonstrates how endogenous pathways can be used for the development of companion therapy in CF.


Asunto(s)
Sistemas de Transporte de Aminoácidos/fisiología , Arginina/metabolismo , Bronquios/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/fisiología , Fibrosis Quística/terapia , Sistemas de Transporte de Aminoácidos/antagonistas & inhibidores , Sistemas de Transporte de Aminoácidos/genética , Transporte Biológico , Bronquios/citología , Células Cultivadas , Fibrosis Quística/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/deficiencia , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Genes Reporteros , Humanos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo II/metabolismo , Proteínas Recombinantes/metabolismo , Propiedades de Superficie , Transducción Genética , Triptófano/análogos & derivados , Triptófano/farmacología
16.
Am J Respir Cell Mol Biol ; 61(6): 747-754, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31184507

RESUMEN

Cystic fibrosis is an autosomal-recessive disease that is caused by a mutant CFTR (cystic fibrosis transmembrane conductance regulator) gene and is characterized by chronic bacterial lung infections and inflammation. Complementation with functional CFTR normalizes anion transport across the airway surface. Adeno-associated virus (AAV) is a useful vector for gene therapy because of its low immunogenicity and ability to persist for months to years. However, because its episomal expression may decrease after cell division, readministration of the AAV vector may be required. To overcome this, we designed an integrating AAV-based CFTR-expressing vector, termed piggyBac (PB)/AAV, carrying CFTR flanked by the terminal repeats of the piggyBac transposon. With codelivery of the piggyBac transposase, PB/AAV can integrate into the host genome. Because of the packaging constraints of AAV, careful consideration was required to ensure that the vector would package and express its CFTR cDNA cargo. In this short-term study, PB/AAV-CFTR was aerosolized to the airways of CF pigs in the absence of the transposase. Two weeks later, transepithelial Cl- current was restored in freshly excised tracheal and bronchial tissue. Additionally, we observed an increase in tracheal airway surface liquid pH and bacterial killing in comparison with untreated CF pigs. Airway surface liquid from primary airway cells cultured from treated CF pigs exhibited increased pH correlating with decreased viscosity. Together, these results show that complementing CFTR in CF pigs with PB/AAV rescues the anion transport defect in a large-animal CF model. Delivery of this integrating viral vector system to airway progenitor cells could lead to persistent, life-long expression in vivo.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/uso terapéutico , Fibrosis Quística/terapia , Dependovirus/genética , Terapia Genética , Vectores Genéticos/uso terapéutico , Animales , Animales Recién Nacidos , Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/administración & dosificación , Regulador de Conductancia de Transmembrana de Fibrosis Quística/deficiencia , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Elementos Transponibles de ADN , Técnicas de Inactivación de Genes , Genes Sintéticos , Humanos , Regiones Promotoras Genéticas , Staphylococcus aureus , Porcinos , Tráquea/metabolismo , Tráquea/microbiología , Integración Viral
17.
Nature ; 567(7748): 405-408, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30867598

RESUMEN

Loss-of-function mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) compromise epithelial HCO3- and Cl- secretion, reduce airway surface liquid pH, and impair respiratory host defences in people with cystic fibrosis1-3. Here we report that apical addition of amphotericin B, a small molecule that forms unselective ion channels, restored HCO3- secretion and increased airway surface liquid pH in cultured airway epithelia from people with cystic fibrosis. These effects required the basolateral Na+, K+-ATPase, indicating that apical amphotericin B channels functionally interfaced with this driver of anion secretion. Amphotericin B also restored airway surface liquid pH, viscosity, and antibacterial activity in primary cultures of airway epithelia from people with cystic fibrosis caused by different mutations, including ones that do not yield CFTR, and increased airway surface liquid pH in CFTR-null pigs in vivo. Thus, unselective small-molecule ion channels can restore host defences in cystic fibrosis airway epithelia via a mechanism that is independent of CFTR and is therefore independent of genotype.


Asunto(s)
Fibrosis Quística/metabolismo , Epitelio/metabolismo , Canales Iónicos/metabolismo , Mucosa Respiratoria/metabolismo , Sistema Respiratorio/metabolismo , Anfotericina B/farmacología , Animales , Bicarbonatos/metabolismo , Células Cultivadas , Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/deficiencia , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Epitelio/efectos de los fármacos , Femenino , Humanos , Concentración de Iones de Hidrógeno , Masculino , Mucosa Respiratoria/efectos de los fármacos , Sistema Respiratorio/efectos de los fármacos , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Porcinos
18.
Dig Dis Sci ; 64(9): 2527-2537, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-30874987

RESUMEN

BACKGROUND: Although ClC-2 channels are important in colonic Cl- secretion, it is unclear about their roles in small intestinal anion secretion. Therefore, we sought to examine whether ClC-2 channels play important roles in anion secretion, particularly duodenal bicarbonate secretion (DBS). METHODS: Duodenal mucosae from mice were stripped of seromuscular layers and mounted in Ussing chambers. Both duodenal short-circuit current (Isc) and HCO3- secretion in vitro were simultaneously recorded. DBS in vivo was measured by a CO2-sensitive electrode. RESULTS: Lubiprostone, a selective ClC-2 activator, concentration-dependently increased both duodenal Isc and DBS only when applied basolaterally, but not when applied apically. Removal of extracellular Cl- abolished lubiprostone-induced duodenal Isc, but did not alter HCO3- secretion even in the presence of DIDS, a Cl-/HCO3- exchanger inhibitor. However, further addition of glibenclamide, a CFTR channel blocker, abolished lubiprostone-evoked HCO3- secretion. Moreover, lubiprostone-induced HCO3- secretion was impaired in CFTR-/- mice compared to wild-type littermates. Luminal perfusion of duodenal lumen with lubiprostone did not alter basal DBS in vivo, but lubiprostone (i.p.) was able to induce DBS, which was also significantly inhibited by Cd2+, a ClC-2 channel blocker. [Ca2+]cyt level, Ca2+-activated K+ channel- and cAMP-mediated duodenal Isc, and HCO3- secretion were unchanged by lubiprostone. CONCLUSIONS: We have provided the first evidence for the novel functional role of basolateral ClC-2 channels in the regulation of duodenal anion secretion.


Asunto(s)
Canales de Cloruro/fisiología , Duodeno/metabolismo , Mucosa Intestinal/metabolismo , Animales , Canales de Cloruro CLC-2 , Agonistas de los Canales de Cloruro/farmacología , Canales de Cloruro/antagonistas & inhibidores , Regulador de Conductancia de Transmembrana de Fibrosis Quística/deficiencia , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Duodeno/efectos de los fármacos , Mucosa Intestinal/efectos de los fármacos , Lubiprostona/farmacología , Ratones , Ratones Noqueados
19.
Med Mycol ; 57(Supplement_2): S118-S126, 2019 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-30816976

RESUMEN

Cystic fibrosis (CF), caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene, is the most common inherited life-limiting disease in North European people affecting 90,000 people worldwide. Progressive lung damage caused by recurrent infection and chronic airway inflammation is the major determinant of survival with a median age at death of 29 years. Approximately 60% of CF patients are infected with Aspergillus fumigatus, a ubiquitous environmental fungus, and its presence has been associated with accelerated lung function decline. Half of the patients infected with Aspergillus are <18 years of age. Yet time of acquisition of this fungus and determinants of CF-related Aspergillus disease severity and progression are not known. CFTR expression has been demonstrated in cells of the innate and adaptive immune system and has shown to be critical for normal function. Research delineating the role of CFTR-deficient phagocytes in Aspergillus persistence and infection in the CF lung, has only recently received attention. In this concise review we aim to present the current understanding with respect to when people with CF acquire infection with A. fumigatus and antifungal immune responses by CF immune cells.


Asunto(s)
Aspergillus fumigatus/inmunología , Fibrosis Quística/complicaciones , Inmunidad Innata , Leucocitos/inmunología , Aspergilosis Pulmonar/inmunología , Aspergilosis Pulmonar/patología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/deficiencia , Humanos
20.
Biochim Biophys Acta Mol Basis Dis ; 1865(6): 1323-1331, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30716472

RESUMEN

BACKGROUND: We analyzed the CFTR response to VX-809/VX-770 drugs in conditionally reprogrammed cells (CRC) of human nasal epithelium (HNE) from F508del/F508del patients based on SNP rs7512462 in the Solute Carrier Family 26, Member 9 (SLC26A9; MIM: 608481) gene. METHODS: The Isc-eq measurements of primary nasal epithelial cells from F508del/F508del patients (n = 12) for CFTR function were performed in micro Ussing chambers and compared with non-CF controls (n = 2). Data were analyzed according to the rs7512462 genotype which were determined by real-time PCR. RESULTS: The CRC-HNE cells from F508del/F508del patients evidenced high variability in the basal levels of CFTR function. Also, the rs7512462*C allele showed an increased basal CFTR function and higher responses to VX-809 + VX-770. The rs7512462*CC + CT genotypes together evidenced CFTR function levels of 14.89% relatively to wt/wt (rs7512462*CT alone-15.29%) i.e., almost double of rs7512462*TT (7.13%). Furthermore, sweat [Cl-] and body mass index of patients also evidenced an association with the rs7512462 genotype. CONCLUSION: The CFTR function can be performed in F508del/F508del patient-derived CRC-HNEs and its function and responses to VX-809 + VX-770 combination as well as clinical data, are all associated with the rs7512462 variant, which partially sheds light on the generally inter-individual phenotypic variability and in personalized responses to CFTR modulator drugs.


Asunto(s)
Aminofenoles/farmacología , Aminopiridinas/farmacología , Antiportadores/genética , Benzodioxoles/farmacología , Agonistas de los Canales de Cloruro/farmacología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Células Epiteliales/efectos de los fármacos , Quinolonas/farmacología , Transportadores de Sulfato/genética , Alelos , Antiportadores/metabolismo , Secuencia de Bases , Índice de Masa Corporal , Estudios de Casos y Controles , Reprogramación Celular , Fibrosis Quística/genética , Fibrosis Quística/metabolismo , Fibrosis Quística/patología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/deficiencia , Cámaras de Difusión de Cultivos , Células Epiteliales/metabolismo , Células Epiteliales/patología , Expresión Génica , Genotipo , Humanos , Modelos Biológicos , Mucosa Nasal/efectos de los fármacos , Mucosa Nasal/metabolismo , Mucosa Nasal/patología , Polimorfismo de Nucleótido Simple , Cultivo Primario de Células , Eliminación de Secuencia , Transportadores de Sulfato/metabolismo , Sudor/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...