Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Nucleic Acids Res ; 52(7): 4002-4020, 2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38321934

RESUMEN

Poly(ADP-ribosylation) (PARylation) is a post-translational modification mediated by a subset of ADP-ribosyl transferases (ARTs). Although PARylation-inhibition based therapies are considered as an avenue to combat debilitating diseases such as cancer and myopathies, the role of this modification in physiological processes such as cell differentiation remains unclear. Here, we show that Tankyrase1 (TNKS1), a PARylating ART, plays a major role in myogenesis, a vital process known to drive muscle fiber formation and regeneration. Although all bona fide PARPs are expressed in muscle cells, experiments using siRNA-mediated knockdown or pharmacological inhibition show that TNKS1 is the enzyme responsible of catalyzing PARylation during myogenesis. Via this activity, TNKS1 controls the turnover of mRNAs encoding myogenic regulatory factors such as nucleophosmin (NPM) and myogenin. TNKS1 mediates these effects by targeting RNA-binding proteins such as Human Antigen R (HuR). HuR harbors a conserved TNKS-binding motif (TBM), the mutation of which not only prevents the association of HuR with TNKS1 and its PARylation, but also precludes HuR from regulating the turnover of NPM and myogenin mRNAs as well as from promoting myogenesis. Therefore, our data uncover a new role for TNKS1 as a key modulator of RBP-mediated post-transcriptional events required for vital processes such as myogenesis.


Asunto(s)
Desarrollo de Músculos , Fibras Musculares Esqueléticas , Miogenina , ARN Mensajero , Tanquirasas , Tanquirasas/metabolismo , Tanquirasas/genética , Humanos , ARN Mensajero/metabolismo , ARN Mensajero/genética , Desarrollo de Músculos/genética , Animales , Fibras Musculares Esqueléticas/metabolismo , Ratones , Miogenina/genética , Miogenina/metabolismo , Nucleofosmina , Proteína 1 Similar a ELAV/metabolismo , Proteína 1 Similar a ELAV/genética , Estabilidad del ARN/genética , Poli ADP Ribosilación/genética , Línea Celular , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , Diferenciación Celular/genética , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética , Células HEK293
2.
Am J Physiol Cell Physiol ; 323(5): C1374-C1392, 2022 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-36121129

RESUMEN

Chronic obstructive pulmonary disease (COPD) is a leading cause of death and cigarette smoke is the main risk factor. Detecting its earliest stages and preventing a decline in lung function are key goals. The pathogenesis of COPD is complex but has some similarities to cystic fibrosis (CF), a disease caused by mutations in the cftr gene. CF leads to chronic inflammation, abnormal mucus, and cycles of infection. Cigarette smoke exposure also causes CFTR dysfunction, and it is probably not a coincidence that inflammation, mucus obstruction, and infections are also characteristics of COPD, although the exacerbations can be quite different. We review here the acute effects of cigarette smoke on CFTR function and its potential role in COPD. Understanding CFTR regulation by cigarette smoke may identify novel drug targets and facilitate the development of therapeutics that reduce the progression and severity of COPD.


Asunto(s)
Fumar Cigarrillos , Fibrosis Quística , Enfermedad Pulmonar Obstructiva Crónica , Humanos , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Fumar Cigarrillos/efectos adversos , Enfermedad Pulmonar Obstructiva Crónica/genética , Fibrosis Quística/genética , Nicotiana , Inflamación
3.
Nat Commun ; 13(1): 3586, 2022 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-35739107

RESUMEN

Impaired activity of the chloride channel CFTR is the cause of cystic fibrosis. 14-3-3 proteins have been shown to stabilize CFTR and increase its biogenesis and activity. Here, we report the identification and mechanism of action of a macrocycle stabilizing the 14-3-3/CFTR complex. This molecule rescues plasma membrane localization and chloride transport of F508del-CFTR and works additively with the CFTR pharmacological chaperone corrector lumacaftor (VX-809) and the triple combination Trikafta®. This macrocycle is a useful tool to study the CFTR/14-3-3 interaction and the potential of molecular glues in cystic fibrosis therapeutics.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística , Fibrosis Quística , Aminofenoles/metabolismo , Aminopiridinas/metabolismo , Aminopiridinas/farmacología , Membrana Celular/metabolismo , Fibrosis Quística/tratamiento farmacológico , Fibrosis Quística/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Humanos , Mutación
4.
Sci Rep ; 12(1): 4595, 2022 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-35302062

RESUMEN

Most cases of cystic fibrosis (CF) are caused by class 2 mutations in the cystic fibrosis transmembrane regulator (CFTR). These proteins preserve some channel function but are retained in the endoplasmic reticulum (ER). Partial rescue of the most common CFTR class 2 mutant, F508del-CFTR, has been achieved through the development of pharmacological chaperones (Tezacaftor and Elexacaftor) that bind CFTR directly. However, it is not clear whether these drugs will rescue all class 2 CFTR mutants to a medically relevant level. We have previously shown that the nonsteroidal anti-inflammatory drug (NSAID) ibuprofen can correct F508del-CFTR trafficking. Here, we utilized RNAi and pharmacological inhibitors to determine the mechanism of action of the NSAID glafenine. Using cellular thermal stability assays (CETSAs), we show that it is a proteostasis modulator. Using medicinal chemistry, we identified a derivative with a fourfold increase in CFTR corrector potency. Furthermore, we show that these novel arachidonic acid pathway inhibitors can rescue difficult-to-correct class 2 mutants, such as G85E-CFTR > 13%, that of non-CF cells in well-differentiated HBE cells. Thus, the results suggest that targeting the arachidonic acid pathway may be a profitable way of developing correctors of certain previously hard-to-correct class 2 CFTR mutations.


Asunto(s)
Fibrosis Quística , Glafenina , Antiinflamatorios no Esteroideos/farmacología , Antiinflamatorios no Esteroideos/uso terapéutico , Ácido Araquidónico , Ciclooxigenasa 2/metabolismo , Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Glafenina/uso terapéutico , Humanos , Mutación
5.
Cells ; 11(1)2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-35011698

RESUMEN

Although some therapeutic progress has been achieved in developing small molecules that correct F508del-CFTR defects, the mechanism of action (MoA) of these compounds remain poorly elucidated. Here, we investigated the effects and MoA of MCG1516A, a newly developed F508del-CFTR corrector. MCG1516A effects on wild-type (WT) and F508del-CFTR were assessed by immunofluorescence microscopy, and biochemical and functional assays both in cell lines and in intestinal organoids. To shed light on the MoA of MCG1516A, we evaluated its additivity to the FDA-approved corrector VX-661, low temperature, genetic revertants of F508del-CFTR (G550E, R1070W, and 4RK), and the traffic-null variant DD/AA. Finally, we explored the ability of MCG1516A to rescue trafficking and function of other CF-causing mutations. We found that MCG1516A rescues F508del-CFTR with additive effects to VX-661. A similar behavior was observed for WT-CFTR. Under low temperature incubation, F508del-CFTR demonstrated an additivity in processing and function with VX-661, but not with MCG1516A. In contrast, both compounds promoted additional effects to low temperature to WT-CFTR. MCG1516A demonstrated additivity to genetic revertant R1070W, while VX-661 was additive to G550E and 4RK. Nevertheless, none of these compounds rescued DD/AA trafficking. Both MCG1516A and VX-661 rescued CFTR processing of L206W- and R334W-CFTR with greater effects when these compounds were combined. In summary, the absence of additivity of MCG1516A to genetic revertant G550E suggests a putative binding site for this compound on NBD1:NBD2 interface. Therefore, a combination of MCG1516A with compounds able to rescue DD/AA traffic, or mimicking the actions of revertant R1070W (e.g., VX-661), could enhance correction of F508del-CFTR defects.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Fibrosis Quística/genética , Descubrimiento de Drogas/métodos , Humanos , Mutación , Pliegue de Proteína
6.
ACS Omega ; 5(40): 25593-25604, 2020 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-33073085

RESUMEN

The marine natural product latonduine A (1) shows F508del-cystic fibrosis transmembrane regulator (CFTR) corrector activity in cell-based assays. Pull-down experiments, enzyme inhibition assays, and siRNA knockdown experiments suggest that the F508del-CFTR corrector activities of latonduine A and a synthetic analogue MCG315 (4) result from simultaneous inhibition of PARP3 and PARP16. A library of synthetic latonduine A analogs has been prepared in an attempt to separate the PARP3 and PARP16 inhibitory properties of latonduine A with the goal of discovering selective small-molecule PARP3 and PARP16 inhibitory cell biology tools that could confirm the proposed dual-target F508del-CFTR corrector mechanism of action. The structure activity relationship (SAR) study reported herein has resulted in the discovery of the modestly potent (IC50 3.1 µM) PARP3 selective inhibitor (±)-5-hydroxy-4-phenyl-2,3,4,5-tetrahydro-1H-benzo[c]azepin-1-one (5) that shows 96-fold greater potency for inhibition of PARP3 compared with its inhibition of PARP16 in vitro and the potent (IC50 0.362 µM) PARP16 selective inhibitor (±)-7,8-dichloro-5-hydroxy-4-(pyridin-2-yl)-2,3,4,5-tetrahydro-1H-benzo[c]azepin-1-one (6) that shows 205-fold selectivity for PARP16 compared with PARP3 in vitro. At 1 or 10 µM, neither 5 or 6 alone showed F508del-CFTR corrector activity, but when added together at 1 or 10 µM each, the combination exhibited F508del-CFTR corrector activity identical to 1 or 10 µM latonduine A (1), respectively, supporting its novel dual PARP target mechanism of action. Latonduine A (1) showed additive in vitro corrector activity in combination with the clinically approved corrector VX809, making it a potential new partner for cystic fibrosis combination drug therapies.

7.
Biochem Pharmacol ; 180: 114133, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32628927

RESUMEN

Despite progress in developing pharmacotherapies to rescue F508del-CFTR, the most prevalent Cystic Fibrosis (CF)-causing mutation, individuals homozygous for this mutation still face several disease-related symptoms. Thus, more potent compound combinations are still needed. Here, we investigated the mechanism of action (MoA) of RDR01752, a novel F508del-CFTR trafficking corrector. F508del-CFTR correction by RDR01752 was assessed by biochemical, immunofluorescence microscopy and functional assays in cell lines and in intestinal organoids. To determine the MoA of RDR01752, we assessed its additive effects to those of genetic revertants of F508del-CFTR, the FDA-approved corrector drugs VX-809 and VX-661, and low temperature. Our data demonstrated that RDR01752 rescues F508del-CFTR processing and plasma membrane (PM) expression to similar levels of VX-809 in cell lines, although RDR01752 produced lower functional rescue. However, in functional assays using intestinal organoids (F508del/F508del), RDR01752, VX-809 and VX-661 had similar efficacy. RDR01752 demonstrated additivity to revertants 4RK and G550E, but not to R1070W, as previously shown for VX-809. RDR01752 was also additive to low temperature. Co-treatment of RDR01752 and VX-809 did not increase F508del-CFTR PM expression and function compared to each corrector alone. The lack of additivity of RDR01752 with the genetic revertant R1070W suggests that this compound has the same effect as the insertion of tryptophan at 1070, i.e., filling the pocket at the NBD1:ICL4 interface in F508del-CFTR, similarly to VX-809. Combination of RDR01752 with correctors mimicking the rescue by revertants G550E or 4RK could thus maximize rescue of F508del-CFTR.


Asunto(s)
Aminopiridinas/farmacología , Benzodioxoles/farmacología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Fibrosis Quística/tratamiento farmacológico , Fibrosis Quística/genética , Indoles/farmacología , Bronquios/efectos de los fármacos , Bronquios/metabolismo , Línea Celular , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Descubrimiento de Drogas , Humanos , Mutación , Organoides/efectos de los fármacos , Organoides/metabolismo , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/genética
8.
Expert Opin Ther Targets ; 23(8): 711-724, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31169041

RESUMEN

Introduction: Cystic fibrosis (CF) is the most frequent lethal orphan disease and is caused by mutations in the CFTR gene. The most frequent mutation F508del-CFTR affects multiple organs; infections and subsequent infections and complications in the lung lead to death. Areas covered: This review focuses on new targets and mechanisms that are attracting interest for the development of CF therapies. The F508del-CFTR protein is retained in the endoplasmic reticulum (ER) but has some function if it can traffic to the plasma membrane. Cell-based assays have been used to screen chemical libraries for small molecule correctors that restore its trafficking. Pharmacological chaperones are correctors that bind directly to the F508del-CFTR mutant and promote its folding and trafficking. Other correctors fall into a heterogeneous class of proteostasis modulators that act indirectly by altering cellular homeostasis. Expert opinion: Pharmacological chaperones have so far been the most successful correctors of F508del-CFTR trafficking, but their level of correction means that more than one corrector is required. Proteostasis modulators have low levels of correction but hold promise because some can correct several different CFTR mutations. Identification of their cellular targets and the potential for development may lead to new therapies for CF.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Fibrosis Quística/tratamiento farmacológico , Terapia Molecular Dirigida , Animales , Fibrosis Quística/genética , Fibrosis Quística/fisiopatología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Desarrollo de Medicamentos , Retículo Endoplásmico/metabolismo , Humanos , Mutación , Pliegue de Proteína
9.
Sci Rep ; 8(1): 11404, 2018 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-30061653

RESUMEN

Pharmacological chaperones (e.g. VX-809, lumacaftor) that bind directly to F508del-CFTR and correct its mislocalization are promising therapeutics for Cystic Fibrosis (CF). However to date, individual correctors provide only ~4% improvement in lung function measured as FEV1, suggesting that multiple drugs will be needed to achieve substantial clinical benefit. Here we examine if multiple sites for pharmacological chaperones exist and can be targeted to enhance the rescue of F508del-CFTR with the premise that additive or synergistic rescue by multiple pharmacological chaperones compared to single correctors indicates that they have different sites of action. First, we found that a combination of the pharmacological chaperones VX-809 and RDR1 provide additive correction of F508del-CFTR. Then using cellular thermal stability assays (CETSA) we demonstrated the possibility of a third pharmacologically important site using the novel pharmacological chaperone tool compound 4-methyl-N-[3-(morpholin-4-yl) quinoxalin-2-yl] benzenesulfonamide (MCG1516A). All three pharmacological chaperones appear to interact with the first nucleotide-binding domain (NBD1). The triple combination of MCG1516A, RDR1, and VX-809 restored CFTR function to >20% that of non-CF cells in well differentiated HBE cells and to much higher levels in other cell types. Thus the results suggest the presence of at least three distinct sites for pharmacological chaperones on F508del-CFTR NBD1, encouraging the development of triple corrector combinations.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Aminopiridinas/farmacología , Animales , Benzodioxoles/farmacología , Línea Celular , Polaridad Celular/efectos de los fármacos , Sinergismo Farmacológico , Quimioterapia Combinada , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Humanos , Potenciales de la Membrana/efectos de los fármacos , Mutación/genética , Dominios Proteicos
10.
Curr Opin Pharmacol ; 34: 105-111, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-29080476

RESUMEN

These are exciting times in the development of therapeutics for cystic fibrosis (CF). New correctors and potentiators of the cystic fibrosis transmembrane conductance regulator (CFTR) are being developed in academic laboratories and pharmaceutical companies, and the field is just beginning to understand their mechanisms of action. Studies of CFTR modulators are also yielding insight into the general principles and strategies that can be used when developing pharmacological chaperones, a new class of drugs. Combining two or even three correctors with a potentiator is an especially promising approach which should lead to further improvements in efficacy and clinical benefit for patients.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Fibrosis Quística/tratamiento farmacológico , Aminofenoles/farmacología , Sitios de Unión , Agonistas de los Canales de Cloruro/farmacología , Agonistas de los Canales de Cloruro/uso terapéutico , Fibrosis Quística/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Quimioterapia Combinada , Humanos , Mutación , Quinolonas/farmacología
11.
Mol Pharmacol ; 90(2): 65-79, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27193581

RESUMEN

Cystic fibrosis (CF) is a major lethal genetic disease caused by mutations in the CF transmembrane conductance regulator gene (CFTR). This encodes a chloride ion channel on the apical surface of epithelial cells. The most common mutation in CFTR (F508del-CFTR) generates a protein that is misfolded and retained in the endoplasmic reticulum. Identifying small molecules that correct this CFTR trafficking defect is a promising approach in CF therapy. However, to date only modest efficacy has been reported for correctors in clinical trials. We identified the marine sponge metabolite latonduine as a corrector. We have now developed a series of latonduine derivatives that are more potent F508del-CFTR correctors with one (MCG315 [2,3-dihydro-1H-2-benzazepin-1-one]) having 10-fold increased corrector activity and an EC50 of 72.25 nM. We show that the latonduine analogs inhibit poly-ADP ribose polymerase (PARP) isozymes 1, 3, and 16. Further our molecular modeling studies point to the latonduine analogs binding to the PARP nicotinamide-binding domain. We established the relationship between the ability of the latonduine analogs to inhibit PARP-16 and their ability to correct F508del-CFTR trafficking. We show that latonduine can inhibit both PARP-3 and -16 and that this is necessary for CFTR correction. We demonstrate that latonduine triggers correction by regulating the activity of the unfolded protein response activator inositol-requiring enzyme (IRE-1) via modulation of the level of its ribosylation by PARP-16. These results establish latonduines novel site of action as well as its proteostatic mechanism of action.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Compuestos Heterocíclicos con 3 Anillos/farmacología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Adenosina Difosfato Ribosa/metabolismo , Animales , Proteínas de Ciclo Celular/química , Línea Celular , Endorribonucleasas/metabolismo , Técnicas de Silenciamiento del Gen , Glicoproteínas/metabolismo , Compuestos Heterocíclicos con 3 Anillos/química , Humanos , Modelos Moleculares , Inhibidores de Poli(ADP-Ribosa) Polimerasas/química , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Poli(ADP-Ribosa) Polimerasas/química , Transporte de Proteínas/efectos de los fármacos , ARN Interferente Pequeño/metabolismo , Respuesta de Proteína Desplegada/efectos de los fármacos
12.
Proc Natl Acad Sci U S A ; 113(9): E1152-61, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26888287

RESUMEN

Cystic fibrosis is a fatal genetic disease, most frequently caused by the retention of the CFTR (cystic fibrosis transmembrane conductance regulator) mutant protein in the endoplasmic reticulum (ER). The binding of the 14-3-3 protein to the CFTR regulatory (R) domain has been found to enhance CFTR trafficking to the plasma membrane. To define the mechanism of action of this protein-protein interaction, we have examined the interaction in vitro. The disordered multiphosphorylated R domain contains nine different 14-3-3 binding motifs. Furthermore, the 14-3-3 protein forms a dimer containing two amphipathic grooves that can potentially bind these phosphorylated motifs. This results in a number of possible binding mechanisms between these two proteins. Using multiple biochemical assays and crystal structures, we show that the interaction between them is governed by two binding sites: The key binding site of CFTR (pS768) occupies one groove of the 14-3-3 dimer, and a weaker, secondary binding site occupies the other binding groove. We show that fusicoccin-A, a natural-product tool compound used in studies of 14-3-3 biology, can stabilize the interaction between 14-3-3 and CFTR by selectively interacting with a secondary binding motif of CFTR (pS753). The stabilization of this interaction stimulates the trafficking of mutant CFTR to the plasma membrane. This definition of the druggability of the 14-3-3-CFTR interface might offer an approach for cystic fibrosis therapeutics.


Asunto(s)
Proteínas 14-3-3/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Proteínas 14-3-3/química , Secuencia de Aminoácidos , Sitios de Unión , Calorimetría , Regulador de Conductancia de Transmembrana de Fibrosis Quística/química , Modelos Moleculares , Datos de Secuencia Molecular
13.
Br J Pharmacol ; 173(3): 459-70, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26492939

RESUMEN

BACKGROUND AND PURPOSE: The most common cystic fibrosis (CF) mutation F508del inhibits the gating and surface expression of CFTR, a plasma membrane anion channel. Optimal pharmacotherapies will probably require both a 'potentiator' to increase channel open probability and a 'corrector' that improves folding and trafficking of the mutant protein and its stability at the cell surface. Interaction between CF drugs has been reported but remains poorly understood. EXPERIMENTAL APPROACH: CF bronchial epithelial cells were exposed to the corrector VX-809 (lumacaftor) and potentiator VX-770 (ivacaftor) individually or in combination. Functional expression of CFTR was assayed as the forskolin-stimulated short-circuit current (Isc ) across airway epithelial monolayers expressing F508del CFTR. KEY RESULTS: The potentiated Isc response during forskolin stimulation was increased sixfold after pretreatment with VX-809 alone and reached ~11% that measured across non-CF monolayers. VX-770 (100 nM) and genistein (50 µM) caused similar levels of potentiation, which were not additive and were abolished by the CFTR inhibitor CFTRinh -172. The unbound fraction of VX-770 in plasma was 0.13 ± 0.04%, which together with previous measurements in patients given 250 mg p.o. twice daily, suggests a peak free plasma concentration of 1.5-8.5 nM. Chronic exposure to high VX-770 concentrations (>1 µM) inhibited functional correction by VX-809 but not in the presence of physiological protein levels (20-40 mg·mL(-1) ). Chronic exposure to a low concentration of VX-770 (100 nM) together with VX-809 (1 µM) also did not reduce the forskolin-stimulated Isc , relative to cells chronically exposed to VX-809 alone, provided it was assayed acutely using the same, clinically relevant concentration of potentiator. CONCLUSIONS AND IMPLICATIONS: Chronic exposure to clinically relevant concentrations of VX-770 did not reduce F508del CFTR function. Therapeutic benefit of VX-770 + VX-809 (Orkambi) is probably limited by the efficacy of VX-809 rather than by inhibition by VX-770.


Asunto(s)
Aminofenoles/farmacología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Quinolonas/farmacología , Aminopiridinas/farmacología , Benzodioxoles/farmacología , Bronquios/citología , Línea Celular , Células Cultivadas , Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Interacciones Farmacológicas , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Humanos , Mutación
14.
Elife ; 42015 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-26701908

RESUMEN

Cystic fibrosis (CF) is caused by mutations in CF transmembrane conductance regulator (CFTR). The most frequent mutation (F508del-CFTR) results in altered proteostasis, that is, in the misfolding and intracellular degradation of the protein. The F508del-CFTR proteostasis machinery and its homeostatic regulation are well studied, while the question whether 'classical' signalling pathways and phosphorylation cascades might control proteostasis remains barely explored. Here, we have unravelled signalling cascades acting selectively on the F508del-CFTR folding-trafficking defects by analysing the mechanisms of action of F508del-CFTR proteostasis regulator drugs through an approach based on transcriptional profiling followed by deconvolution of their gene signatures. Targeting multiple components of these signalling pathways resulted in potent and specific correction of F508del-CFTR proteostasis and in synergy with pharmacochaperones. These results provide new insights into the physiology of cellular proteostasis and a rational basis for developing effective pharmacological correctors of the F508del-CFTR defect.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Fibrosis Quística/genética , Deficiencias en la Proteostasis/genética , Transducción de Señal , Línea Celular , Inhibidores Enzimáticos/metabolismo , Perfilación de la Expresión Génica , Humanos , Pliegue de Proteína , Proteolisis , Eliminación de Secuencia
15.
J Pediatr Orthop ; 35(1): 62-8, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24674894

RESUMEN

BACKGROUND: Early recognition of failure of reduction during abduction splinting in developmental dysplasia of the hip (DDH) is essential to avoid inappropriate prolonged damaging harness use. Graf ultrasound scanning (USS) is an accepted gold standard for assessing neonatal hip dysplasia but cannot verify reduction in the abducted position of a neonatal hip in a Pavlik harness. We describe a new anatomic landmark of the hip using anterior USS-the "ischial limb," correlate this to the position of the femoral head in reduced and dislocated positions, and assess the application of this in the assessment of DDH. METHODS: A porcine model was used to identify anatomic, radiologic, and ultrasonic features comparable to the human neonatal hip. A pilot clinical study was conducted of 50 neonates (11M:39F) treated for DDH in Pavlik harness, utilizing weekly anterior USS to determine the concentricity of femoral head reduction relative to the ischial limb and whether this could identify any failures to achieve reduction. RESULTS: The animal model identified an ultrasound landmark, termed the ischial limb that represents the ossification zone of the ischial contribution to the tri-radiate acetabular cartilage, and approximates to the center of the acetabulum. Anterior USS clearly demonstrated this ischial limb in clinical practice and its relation to the femoral head in all dysplastic hips (n=79). Immediate concentric reduction was observed in 11 patients, more gradual reduction over 3 weeks in 37 patients, and failure to achieve stable reduction was verified in 2 patients. The technique was reproducible in 100% of patients (n=50). Mean additional clinic time was 5 minutes per visit. CONCLUSIONS: The ischial limb, identified on anterior hip USS, delineates the position of the tri-radiate cartilage. This technique efficiently assesses hip reduction during Pavlik harness treatment and can differentiate between failure to achieve reduction or maintain a stable reduction at an earlier stage than conventional US methods. LEVEL OF EVIDENCE: Development of diagnostic criteria on basis of consecutive patients: level 2 evidence.


Asunto(s)
Acetábulo/diagnóstico por imagen , Desarrollo Infantil/fisiología , Cabeza Femoral/diagnóstico por imagen , Luxación de la Cadera/diagnóstico , Animales , Femenino , Luxación de la Cadera/etiología , Luxación de la Cadera/prevención & control , Humanos , Recién Nacido , Masculino , Modelos Animales , Tamizaje Neonatal/métodos , Aparatos Ortopédicos , Posicionamiento del Paciente/métodos , Proyectos Piloto , Porcinos , Ultrasonografía
16.
J Cyst Fibros ; 14(1): 16-25, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24974227

RESUMEN

BACKGROUND: Small molecules as shown by VX809 can rescue the mislocalization of F508del-CFTR. The aim of this study was to identify correctors with a clinical history and their targets of action. METHODS: CFTR correctors were screened using two F508del-CFTR expressing cell based HTS assays. Electrophysiological studies using CFBE41o(-) and HBE cells and in-vivo mouse assays confirmed CFTR rescue. The target of action was attained using pharmacological inhibitors and siRNA to specific genes. RESULTS: Ibuprofen was identified as a CFTR corrector. Ibuprofen treatment of polarized CFBE41o(-) monolayers increased the short-circuit current (Isc) response to stimulation. In vivo CF mice treatment with ibuprofen restored the CFTR trafficking. SiRNA knock down of cyclooxygenase expression caused partial F508del-CFTR correction. CONCLUSION: These studies show that ibuprofen is a CFTR corrector and that it causes correction by COX-1 inhibition. Hence ibuprofen may be suitable to be part of a future CF combination therapy.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Inhibidores de la Ciclooxigenasa/farmacología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/efectos de los fármacos , Fibrosis Quística/tratamiento farmacológico , Ibuprofeno/farmacología , Animales , Movimiento Celular/genética , Células Cultivadas/efectos de los fármacos , Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Endogámicos CFTR , Mutación , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/genética , ARN Interferente Pequeño/genética , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
17.
J Orthop ; 11(4): 174-9, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25561752

RESUMEN

AIM: Ultrasound scanning (USS) is used for diagnosis and surveillance in developmental dysplasia of the hip (DDH). Lateral coronal scanning is performed with the hip flexed, in neutral adduction. In this position an unstable hip may dislocate, failing to demonstrate a reducible hip, leading to abandonment of harness treatment. Anterior ultrasound permits imaging of the flexed abducted hip in harness. This study evaluates the role of anterior & lateral USS in determining duration of treatment and reduction in DDH. METHOD: Between 1997 & 2010, 233 patients requiring harness treatment received lateral USS, with dislocated & dysplastic hips re-imaged fortnightly. From 2005, anterior USS was used additionally to assess reduction in harness. RESULTS: One-hundred and eighteen patients (167 hips) received lateral USS, 115 (160 hips) received both. In the lateral cohort, 103 (140 hips) were treated successfully, mean duration 66.2 days (95% CI 60.2-72.1), with 15 (26 hips) failures (15.5%), mean 30 (CI 95% 19.3-40.6). In the anterior cohort, 107 (150 hips) were treated successfully, mean 53.3 (95% CI 49.8-56.7), with 8 (10 hips) failures (6.25%), mean 35.3 (CI 95% 25.5-44.9). Children receiving an anterior USS had a shorter duration of treatment (p = 0.011) and no difference in failures (p = 0.21). CONCLUSIONS: A reduced duration of treatment for Graf 3 hips was observed. Anterior ultrasound allows earlier recognition of hips that fail to stabilize, via two observed modes of failure; failure of hip reduction and failure to stabilize after reduction.

18.
Chem Biol ; 19(10): 1288-99, 2012 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-23102222

RESUMEN

Mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene cause CF. The most common mutation, F508 deletion, causes CFTR misfolding and endoplasmic reticulum retention, preventing it from trafficking to the cell surface. One approach to CF treatment is to identify compounds that correct the trafficking defect. We screened a marine extract collection and, after extract, deconvolution identified the latonduines as F508del-CFTR trafficking correctors that give functional correction in vivo. Using a biotinylated azido derivative of latonduine, we identified the poly(ADP-ribose) polymerase (PARP) family as latonduine target proteins. We show that latonduine binds to PARPs 1, 2, 3, 4, 5a, and 5b and inhibits PARP activity, especially PARP-3. Thus, latonduine corrects F508del-CFTR trafficking by modulating PARP activity. Latonduines represent pharmacologic agents for F508del-CFTR correction, and PARP-3 is a pathway for the development of CF treatments.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Compuestos Heterocíclicos con 3 Anillos/farmacología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Transporte de Proteínas/efectos de los fármacos , Alcaloides/química , Alcaloides/farmacología , Animales , Línea Celular , Cricetinae , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Retículo Endoplásmico/metabolismo , Compuestos Heterocíclicos con 3 Anillos/química , Humanos , Ratones , Mutación , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Poli(ADP-Ribosa) Polimerasas/genética , Poríferos/química , Unión Proteica , Pliegue de Proteína/efectos de los fármacos , Interferencia de ARN , ARN Interferente Pequeño/metabolismo
19.
Front Pharmacol ; 3: 176, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23060796

RESUMEN

Most cases of cystic fibrosis (CF) are caused by the deletion of a single phenylalanine residue at position 508 of the cystic fibrosis transmembrane conductance regulator (CFTR). The mutant F508del-CFTR is retained in the endoplasmic reticulum and degraded, but can be induced by low temperature incubation (29°C) to traffic to the plasma membrane where it functions as a chloride channel. Here we show that, cardiac glycosides, at nanomolar concentrations, can partially correct the trafficking of F508del-CFTR in human CF bronchial epithelial cells (CFBE41o-) and in an F508del-CFTR mouse model. Comparison of the transcriptional profiles obtained with polarized CFBE41o-cells after treatment with ouabain and by low temperature has revealed a striking similarity between the two corrector treatments that is not shared with other correctors. In summary, our study shows a novel function of ouabain and its analogs in the regulation of F508del-CFTR trafficking and suggests that compounds that mimic this low temperature correction of trafficking will provide new avenues for the development of therapeutics for CF.

20.
Front Pharmacol ; 3: 165, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22988441

RESUMEN

Most cystic fibrosis is caused by mutations in CFTR that prevent its trafficking from the ER to the plasma membrane and is associated with exaggerated inflammation, altered metabolism, and diminished responses to oxidative stress. PARP-1 is activated by oxidative stress and causes energy depletion and cell dysfunction. Inhibition of this enzyme protects against excessive inflammation and recent studies have also implicated it in intracellular protein trafficking. We hypothesized that PARP-1 activity is altered in CF and affects trafficking and function of the most common CF mutant ΔF508 CFTR. Indeed, PARP-1 activity was 2.9-fold higher in CF (ΔF508/ΔF508) human bronchial epithelial primary cells than in non-CF cells, and similar results were obtained by comparing CF vs. non-CF bronchial epithelial cell lines (2.5-fold higher in CFBE41o(-) vs. 16HBE14o(-), P < 0.002). A PARP-1 inhibitor (ABT-888, Veliparib) partially restored CFTR channel activity in CFBE41o(-) cells overexpressing ΔF508 CFTR. Similarly, reducing PARP-1 activity by 85% in ileum from transgenic CF mice (Cftr(tm1)Eur) partially rescued ΔF508 CFTR activity to 7% of wild type mouse levels, and similar correction (7.8%) was observed in vivo by measuring salivary secretion. Inhibiting PARP-1 with ABT-888 or siRNA partially restored ΔF508 CFTR trafficking in cell lines, and most ΔF508 CFTR was complex glycosylated when heterologously expressed in PARP-1(-/-) mouse embryonic fibroblasts. Finally, levels of the mature glycoform of CFTR were reduced by peroxynitrite, a strong activator of PARP-1. These results demonstrate that PARP-1 activity is increased in CF, and identify a novel pathway that could be targeted by proteostatic correctors of CFTR trafficking.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA