Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
J Med Chem ; 65(7): 5212-5243, 2022 04 14.
Article in English | MEDLINE | ID: mdl-35377645

ABSTRACT

In cystic fibrosis (CF), the deletion of phenylalanine 508 (F508del) in the CF transmembrane conductance regulator (CFTR) leads to misfolding and premature degradation of the mutant protein. These defects can be targeted with pharmacological agents named potentiators and correctors. During the past years, several efforts have been devoted to develop and approve new effective molecules. However, their clinical use remains limited, as they fail to fully restore F508del-CFTR biological function. Indeed, the search for CFTR correctors with different and additive mechanisms has recently increased. Among them, drugs that modulate the CFTR proteostasis environment are particularly attractive to enhance therapy effectiveness further. This Perspective focuses on reviewing the recent progress in discovering CFTR proteostasis regulators, mainly describing the design, chemical structure, and structure-activity relationships. The opportunities, challenges, and future directions in this emerging and promising field of research are discussed, as well.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator , Cystic Fibrosis , Proteostasis , Cystic Fibrosis/drug therapy , Cystic Fibrosis/genetics , Cystic Fibrosis/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/drug effects , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Humans , Mutant Proteins/drug effects , Mutant Proteins/genetics , Mutant Proteins/metabolism , Mutation , Protein Folding/drug effects , Proteostasis/drug effects , Proteostasis/physiology
2.
Biomolecules ; 11(10)2021 09 28.
Article in English | MEDLINE | ID: mdl-34680050

ABSTRACT

Cystic fibrosis (CF) is caused by loss-of-function mutations in the CF transmembrane conductance regulator (CFTR) protein, an anion channel that regulates epithelial surface fluid secretion. The deletion of phenylalanine at position 508 (F508del) is the most common CFTR mutation. F508del CFTR is characterized by folding and trafficking defects, resulting in decreased functional expression of the protein on the plasma membrane. Several classes of small molecules, named correctors, have been developed to rescue defective F508del CFTR. Although individual correctors failed to improve the clinical status of CF patients carrying the F508del mutation, better results were obtained using correctors combinations. These results were obtained according to the premise that the administration of correctors having different sites of action should enhance F508del CFTR rescue. We investigated the putative site of action of an aminoarylthiazole 4-(3-chlorophenyl)-N-(3-(methylthio)phenyl)thiazol-2-amine, named FCG, with proven CFTR corrector activity, and its synergistic effect with the corrector VX809. We found that neither the total expression nor the maturation of WT CFTR transiently expressed in human embryonic kidney 293 cells was influenced by FCG, administrated alone or in combination with VX809. On the contrary, FCG was able to enhance F508del CFTR total expression, and its combination with VX809 provided a further effect, being able to increase not only the total expression but also the maturation of the mutant protein. Analyses on different CFTR domains and groups of domains, heterologously expressed in HEK293 cells, show that NBD2 is necessary for FCG corrector activity. Molecular modelling analyses suggest that FCG interacts with a putative region located into the NBD2, ascribing this molecule to class II correctors. Our study indicates that the continuous development and testing of combinations of correctors targeting different structural and functional defects of mutant CFTR is the best strategy to ensure a valuable therapeutic perspective to a larger cohort of CF patients.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis/drug therapy , Mutant Proteins/genetics , Thiazoles/pharmacology , Aminopyridines/pharmacology , Aminopyridines/therapeutic use , Benzodioxoles/pharmacology , Benzodioxoles/therapeutic use , Cell Membrane/drug effects , Cell Membrane/genetics , Cystic Fibrosis/genetics , Cystic Fibrosis/pathology , Epithelial Cells/drug effects , Gene Expression Regulation/drug effects , HEK293 Cells , Humans , Mutant Proteins/drug effects , Phenylalanine/genetics
3.
PLoS Comput Biol ; 17(9): e1009370, 2021 09.
Article in English | MEDLINE | ID: mdl-34529671

ABSTRACT

Three-dimensional structures of proteins can provide important clues into the efficacy of personalized treatment. We perform a structural analysis of variants within three inherited lysosomal storage disorders, comparing variants responsive to pharmacological chaperone treatment to those unresponsive to such treatment. We find that predicted ΔΔG of mutation is higher on average for variants unresponsive to treatment, in the case of datasets for both Fabry disease and Pompe disease, in line with previous findings. Using both a single decision tree and an advanced machine learning approach based on the larger Fabry dataset, we correctly predict responsiveness of three Gaucher disease variants, and we provide predictions for untested variants. Many variants are predicted to be responsive to treatment, suggesting that drug-based treatments may be effective for a number of variants in Gaucher disease. In our analysis, we observe dependence on a topological feature reporting on contact arrangements which is likely connected to the order of folding of protein residues, and we provide a potential justification for this observation based on steady-state cellular kinetics.


Subject(s)
Fabry Disease/drug therapy , Gaucher Disease/drug therapy , Glycogen Storage Disease Type II/drug therapy , Computational Biology , Decision Trees , Fabry Disease/genetics , Fabry Disease/metabolism , Gaucher Disease/genetics , Gaucher Disease/metabolism , Glycogen Storage Disease Type II/genetics , Glycogen Storage Disease Type II/metabolism , Humans , Kinetics , Machine Learning , Models, Molecular , Mutant Proteins/chemistry , Mutant Proteins/drug effects , Mutant Proteins/genetics , Mutation, Missense , Precision Medicine , Protein Binding , Protein Conformation , Protein Folding , Protein Stability/drug effects , Small Molecule Libraries
4.
J Cyst Fibros ; 20(3): 452-459, 2021 05.
Article in English | MEDLINE | ID: mdl-32674984

ABSTRACT

BACKGROUND: The c.3700A>G mutation, a rare cystic fibrosis (CF)-causing CFTR mutation found mainly in the Middle East, produces full-length transcript encoding a missense mutation (I1234V-CFTR), and a cryptic splice site that deletes 6 amino acids in nucleotide binding domain 2 (I1234del-CFTR). METHODS: FRT cell models expressing I1234V-CFTR and I1234del-CFTR were generated. We also studied an I1234del-CFTR-expressing gene-edited human bronchial (16HBE14o-) cell model, and primary cultures of nasal epithelial cells from a c.3700A>G homozygous subject. To identify improved mutation-specific CFTR modulators, high-throughput screening was done using I1234del-CFTR-expressing FRT cells. Motivated by the in vitro findings, Trikafta was tested in two c.3700A>G homozygous CF subjects. RESULTS: FRT cells expressing full-length I1234V-CFTR had similar function to that of wildtype CFTR. I1234del-CFTR showed reduced activity, with modest activation seen with potentiators VX-770 and GLPG1837, correctors VX-809, VX-661 and VX-445, and low-temperature incubation. Screening identified novel arylsulfonyl-piperazine and spiropiperidine-quinazolinone correctors, which when used in combination with VX-445 increased current ~2-fold compared with the VX-661/VX-445 combination. The combination of VX-770 with arylsulfonamide-pyrrolopyridine, piperidine-pyridoindole or pyrazolo-quinoline potentiators gave 2-4-fold greater current than VX-770 alone. Combination potentiator (co-potentiator) efficacy was also seen in gene-edited I1234del-CFTR-expressing human bronchial epithelial cells. In two CF subjects homozygous for the c.3700A>G mutation, one subject had a 27 mmol/L decrease in sweat chloride and symptomatic improvement on Trikafta, and a second subject showed a small improvement in lung function. CONCLUSIONS: These results support the potential benefit of CFTR modulators, including co-potentiators, for CF caused by the c.3700A>G mutation.


Subject(s)
Chloride Channel Agonists/pharmacology , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis/drug therapy , Cystic Fibrosis/genetics , Mutant Proteins/drug effects , Mutation, Missense , Aminophenols , Aminopyridines , Benzodioxoles , Cells, Cultured , Humans , Indoles , Pyrazoles , Pyridines , Pyrrolidines , Quinolones
5.
Commun Biol ; 3(1): 97, 2020 03 05.
Article in English | MEDLINE | ID: mdl-32139772

ABSTRACT

Mutations to the gene encoding superoxide dismutase-1 (SOD1) were the first genetic elements discovered that cause motor neuron disease (MND). These mutations result in compromised SOD1 dimer stability, with one of the severest and most common mutations Ala4Val (A4V) displaying a propensity to monomerise and aggregate leading to neuronal death. We show that the clinically used ebselen and related analogues promote thermal stability of A4V SOD1 when binding to Cys111 only. We have developed a A4V SOD1 differential scanning fluorescence-based assay on a C6S mutation background that is effective in assessing suitability of compounds. Crystallographic data show that the selenium atom of these compounds binds covalently to A4V SOD1 at Cys111 at the dimer interface, resulting in stabilisation. This together with chemical amenability for hit expansion of ebselen and its on-target SOD1 pharmacological chaperone activity holds remarkable promise for structure-based therapeutics for MND using ebselen as a template.


Subject(s)
Azoles/chemistry , Azoles/pharmacology , Drug Design , Motor Neuron Disease/drug therapy , Organoselenium Compounds/chemistry , Organoselenium Compounds/pharmacology , Superoxide Dismutase-1 , Amino Acid Substitution/genetics , Azoles/chemical synthesis , Azoles/therapeutic use , Crystallography, X-Ray , Drug Discovery/methods , Drug Evaluation, Preclinical/methods , Humans , Isoindoles , Models, Molecular , Molecular Chaperones/chemical synthesis , Molecular Chaperones/chemistry , Molecular Chaperones/therapeutic use , Molecular Docking Simulation , Motor Neuron Disease/genetics , Motor Neuron Disease/metabolism , Motor Neuron Disease/pathology , Mutant Proteins/chemistry , Mutant Proteins/drug effects , Mutant Proteins/genetics , Mutant Proteins/metabolism , Mutation, Missense , Organoselenium Compounds/chemical synthesis , Organoselenium Compounds/isolation & purification , Organoselenium Compounds/therapeutic use , Protein Folding/drug effects , Protein Multimerization/drug effects , Protein Stability/drug effects , Protein Structure, Tertiary , Sulfur Compounds/chemical synthesis , Sulfur Compounds/chemistry , Superoxide Dismutase-1/chemistry , Superoxide Dismutase-1/drug effects , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/metabolism , Thermodynamics
6.
Genome Res ; 30(2): 239-249, 2020 02.
Article in English | MEDLINE | ID: mdl-32051187

ABSTRACT

Understanding the genetic basis for a phenotype is a central goal in biological research. Much has been learnt about bacterial genomes by creating large mutant libraries and looking for conditionally important genes. However, current genome-wide methods are largely unable to assay essential genes which are not amenable to disruption. To overcome this limitation, we developed a new version of "TraDIS" (transposon directed insertion-site sequencing) that we term "TraDIS-Xpress" that combines an inducible promoter into the transposon cassette. This allows controlled overexpression and repression of all genes owing to saturation of inserts adjacent to all open reading frames as well as conventional inactivation. We applied TraDIS-Xpress to identify responses to the biocide triclosan across a range of concentrations. Triclosan is endemic in modern life, but there is uncertainty about its mode of action with a concentration-dependent switch from bacteriostatic to bactericidal action unexplained. Our results show a concentration-dependent response to triclosan with different genes important in survival between static and cidal exposures. These genes include those previously reported to have a role in triclosan resistance as well as a new set of genes, including essential genes. Novel genes identified as being sensitive to triclosan exposure include those involved in barrier function, small molecule uptake, and integrity of transcription and translation. We anticipate the approach we show here, by allowing comparisons across multiple experimental conditions of TraDIS data, and including essential genes, will be a starting point for future work examining how different drug conditions impact bacterial survival mechanisms.


Subject(s)
DNA Transposable Elements/genetics , Genes, Essential/genetics , Genome, Bacterial/drug effects , Triclosan/pharmacology , Escherichia coli/drug effects , Escherichia coli/genetics , Gene Library , Genes, Essential/drug effects , Mutagenesis, Insertional/drug effects , Mutant Proteins/drug effects , Mutant Proteins/genetics , Phenotype
7.
Int J Mol Sci ; 20(21)2019 Nov 01.
Article in English | MEDLINE | ID: mdl-31683989

ABSTRACT

Cystic fibrosis (CF) is a genetic disease associated with the defective function of the cystic fibrosis transmembrane conductance regulator (CFTR) protein that causes obstructive disease and chronic bacterial infections in airway epithelia. The most prevalent CF-causing mutation, the deletion of phenylalanine at position 508 (F508del), leads to CFTR misfolding, trafficking defects and premature degradation. A number of correctors that are able to partially rescue F508del-CFTR processing defects have been identified. Clinical trials have demonstrated that, unfortunately, mono-therapy with the best correctors identified to date does not ameliorate lung function or sweat chloride concentration in homozygous F508del patients. Understanding the mechanisms exerted by currently available correctors to increase mutant F508del-CFTR expression is essential for the development of new CF-therapeutics. We investigated the activity of correctors on the mutant F508del and wild type (WT) CFTR to identify the protein domains whose expression is mostly affected by the action of correctors, and we investigated their mechanisms of action. We found that the four correctors under study, lumacaftor (VX809), the quinazoline derivative VX325, the bithiazole compound corr4a, and the new molecule tezacaftor (VX661), do not influence either the total expression or the maturation of the WT-CFTR transiently expressed in human embryonic kidney 293 (HEK293) cells. Contrarily, they significantly enhance the expression and the maturation of the full length F508del molecule. Three out of four correctors, VX809, VX661 and VX325, seem to specifically improve the expression and the maturation of the mutant CFTR N-half (M1N1, residues 1-633). By contrast, the CFTR C-half (M2N2, residues 837-1480) appears to be the region mainly affected by corr4a. VX809 was shown to stabilize both the WT- and F508del-CFTR N-half isoforms, while VX661 and VX325 demonstrated the ability to enhance the stability only of the mutant F508del polypeptide.


Subject(s)
Aminopyridines/pharmacology , Benzamides/pharmacology , Benzodioxoles/pharmacology , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Indoles/pharmacology , Mutation , Quinazolines/pharmacology , Thiazoles/pharmacology , Cystic Fibrosis/drug therapy , Cystic Fibrosis/genetics , Cystic Fibrosis/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Gene Expression Regulation/drug effects , HEK293 Cells , Humans , Mutant Proteins/drug effects , Mutant Proteins/genetics , Mutant Proteins/metabolism
8.
Neurosci Bull ; 35(6): 1024-1034, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31432317

ABSTRACT

Huntington's disease (HD) is a deadly neurodegenerative disease with abnormal expansion of CAG repeats in the huntingtin gene. Mutant Huntingtin protein (mHTT) forms abnormal aggregates and intranuclear inclusions in specific neurons, resulting in cell death. Here, we tested the ability of a natural heat-shock protein 90 inhibitor, Gedunin, to degrade transfected mHTT in Neuro-2a cells and endogenous mHTT aggregates and intranuclear inclusions in both fibroblasts from HD patients and neurons derived from induced pluripotent stem cells from patients. Our data showed that Gedunin treatment degraded transfected mHTT in Neuro-2a cells, endogenous mHTT aggregates and intranuclear inclusions in fibroblasts from HD patients, and in neurons derived from induced pluripotent stem cells from patients in a dose- and time-dependent manner, and its activity depended on the proteasomal pathway rather than the autophagy route. These findings also showed that although Gedunin degraded abnormal mHTT aggregates and intranuclear inclusions in cells from HD patient, it did not affect normal cells, thus providing a new perspective for using Gedunin to treat HD.


Subject(s)
Huntingtin Protein/drug effects , Huntington Disease/drug therapy , Intranuclear Inclusion Bodies/drug effects , Limonins/pharmacology , Mutant Proteins/drug effects , Protein Aggregates/drug effects , Animals , Cell Culture Techniques , Fibroblasts/drug effects , Heat-Shock Proteins/antagonists & inhibitors , Heat-Shock Proteins/metabolism , Humans , Huntingtin Protein/genetics , Induced Pluripotent Stem Cells/drug effects , Leupeptins/pharmacology , Mice , Mutation , Neurons/drug effects , Proteasome Endopeptidase Complex , Transfection
9.
Br J Haematol ; 187(4): 488-501, 2019 11.
Article in English | MEDLINE | ID: mdl-31309543

ABSTRACT

Mutations in two type-3 receptor tyrosine kinases (RTKs), KIT and FLT3, are common in both acute myeloid leukaemia (AML) and systemic mastocytosis (SM) and lead to hyperactivation of key signalling pathways. A large number of tyrosine kinase inhibitors (TKIs) have been developed that target either FLT3 or KIT and significant clinical benefit has been demonstrated in multiple clinical trials. Given the structural similarity of FLT3 and KIT, it is not surprising that some of these TKIs inhibit both of these receptors. This is typified by midostaurin, which has been approved by the US Food and Drug Administration for mutant FLT3-positive AML and for KIT D816V-positive SM. Here, we compare the in vitro activities of the clinically available FLT3 and KIT inhibitors with those of midostaurin against a panel of cells expressing a variety of oncogenic FLT3 or KIT receptors, including wild-type (wt) FLT3, FLT3-internal tandem duplication (ITD), FLT3 D835Y, the resistance mutant FLT3-ITD+ F691L, KIT D816V, and KIT N822K. We also examined the effects of these inhibitors in vitro and in vivo on cells expressing mutations in c-CBL found in AML that result in hypersensitization of RTKs, such as FLT3 and KIT. The results show a wide spectrum of activity of these various mutations to these clinically available TKIs.


Subject(s)
Antineoplastic Agents/pharmacology , Hematologic Neoplasms/drug therapy , Mutant Proteins/drug effects , Protein Kinase Inhibitors/pharmacology , Aniline Compounds/pharmacology , Aniline Compounds/therapeutic use , Antineoplastic Agents/therapeutic use , Benzimidazoles/pharmacology , Benzimidazoles/therapeutic use , Benzothiazoles/pharmacology , Benzothiazoles/therapeutic use , Cell Line, Tumor , Drug Screening Assays, Antitumor , Hematologic Neoplasms/genetics , Humans , Phenylurea Compounds/pharmacology , Phenylurea Compounds/therapeutic use , Piperidines/pharmacology , Piperidines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-cbl/drug effects , Proto-Oncogene Proteins c-cbl/genetics , Proto-Oncogene Proteins c-kit/drug effects , Proto-Oncogene Proteins c-kit/genetics , Pyrazines/pharmacology , Pyrazines/therapeutic use , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Pyrroles/pharmacology , Pyrroles/therapeutic use , Sorafenib/pharmacology , Sorafenib/therapeutic use , Staurosporine/analogs & derivatives , Staurosporine/pharmacology , Staurosporine/therapeutic use , Triazines/pharmacology , Triazines/therapeutic use , fms-Like Tyrosine Kinase 3/drug effects , fms-Like Tyrosine Kinase 3/genetics
10.
J Cyst Fibros ; 17(5): 595-606, 2018 09.
Article in English | MEDLINE | ID: mdl-29903467

ABSTRACT

BACKGROUND: Current modulator therapies for some cystic fibrosis-causing CFTR mutants, including N1303K, have limited efficacy. We provide evidence here to support combination potentiator (co-potentiator) therapy for mutant CFTRs that are poorly responsive to single potentiators. METHODS: Functional synergy screens done on N1303K and W1282X CFTR, in which small molecules were tested with VX-770, identified arylsulfonamide-pyrrolopyridine, phenoxy-benzimidazole and flavone co-potentiators. RESULTS: A previously identified arylsulfonamide-pyrrolopyridine co-potentiator (ASP-11) added with VX-770 increased N1303K-CFTR current 7-fold more than VX-770 alone. ASP-11 increased by ~65% of the current of G551D-CFTR compared to VX-770, was additive with VX-770 on F508del-CFTR, and activated wild-type CFTR in the absence of a cAMP agonist. ASP-11 efficacy with VX-770 was demonstrated in primary CF human airway cell cultures having N1303K, W1282X and G551D CFTR mutations. Structure-activity studies on 11 synthesized ASP-11 analogs produced compounds with EC50 down to 0.5 µM. CONCLUSIONS: These studies support combination potentiator therapy for CF caused by some CFTR mutations that are not effectively treated by single potentiators.


Subject(s)
Aminophenols/pharmacology , Cystic Fibrosis Transmembrane Conductance Regulator/drug effects , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis/drug therapy , Cystic Fibrosis/genetics , Quinolones/pharmacology , Sulfonamides/pharmacology , Animals , Cell Line , Cells, Cultured , Drug Synergism , Humans , Ion Channel Gating/drug effects , Mutant Proteins/drug effects , Mutation , Structure-Activity Relationship
11.
J Cyst Fibros ; 17(5): 582-594, 2018 09.
Article in English | MEDLINE | ID: mdl-29936070

ABSTRACT

The missing phenylalanine at position 508, located in nucleotide-binding domain (NBD1) of the cystic fibrosis transmembrane regulator (CFTR), is the most common cystic fibrosis mutation. Severe disease-causing mutations also occur in NBD2. To provide information on potential therapeutic strategies for mutations in NBD2, we used a combination of biochemical, cell biological and electrophysiological approaches and newly created cell lines to study two disease-causing NBD2 mutants, N1303K and S1235R. We observed that neither was sensitive to E64, a cysteine protease inhibitor. However, further investigation showed that when treated with a combination of correctors, C4 + C18, both mutants also responded to E64. Further exploration to assess aggresome throughput using the autophagy regulator LC3 as a marker showed that, in the absence of correctors, N1303K showed a stalled throughput of LC3-II to the aggresome. The throughput became active again after treatment with the corrector combination C4 + C18. Confocal microscopic studies showed that the N1303K and S1235R mutant proteins both co-localized with LC3, but this co-localization was abolished by the corrector combination and, to a lesser extent, by VX-809. Both the corrector combination and VX-809 increased the CFTR chloride channel function of both mutants. We conclude that correctors have a dual effect, particularly on N1303K: they improve trafficking and function at the plasma membrane and reduce the association with autophagosomes. After treatment with correctors persistent degradation by the autophagosome may limit restoration of function. Thus, mutations in NBD2 of CFTR, in contrast to ΔF508-CFTR, may require additional personalized strategies to rescue them.


Subject(s)
Aminopyridines/pharmacology , Autophagosomes/physiology , Benzodioxoles/pharmacology , Cystic Fibrosis Transmembrane Conductance Regulator/drug effects , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis/drug therapy , Cystic Fibrosis/genetics , Leucine/analogs & derivatives , Animals , Autophagy , Biological Transport , Blotting, Western , Cell Line , Electric Conductivity , Leucine/pharmacology , Mutant Proteins/drug effects , Mutant Proteins/genetics , Mutation , Small Molecule Libraries/pharmacology
12.
J Cyst Fibros ; 17(5): 573-581, 2018 09.
Article in English | MEDLINE | ID: mdl-29887518

ABSTRACT

BACKGROUND: N1303K, one of the common, severe disease-causing mutations in the CFTR gene, causes both defective biogenesis and gating abnormalities of the CFTR protein. The goals of the present study are to quantitatively assess the gating defects associated with the N1303K mutation and its pharmacological response to CFTR modulators including potentiators VX-770 and GLPG1837 and correctors VX-809, and VX-661. METHODS: Gating behavior and pharmacological responses to CFTR potentiators were assessed using patch-clamp technique in the excised, inside-out mode. We also examined the effects of GLPG1837, VX-770, VX-809 and VX-661 on N1303K-CFTR surface expression using Western blot analysis. RESULTS: Like wild-type (WT) CFTR, N1303K-CFTR channels were activated by protein kinase A-dependent phosphorylation, but the open probability (Po) of phosphorylated N1303K-CFTR was extremely low (~0.03 vs ~0.45 in WT channels). N1303K mutants showed abnormal responses to ATP analogs or mutations that disrupt ATP hydrolysis and/or dimerization of CFTR's two nucleotide-binding domains (NBDs). However, the Po of N1303K-CFTR was dramatically increased by GLPG1837 (~17-fold) and VX-770 (~8-fold). VX-809 or VX-661 enhanced N1303K-CFTR maturation by 2-3 fold, and co-treatment with GLPG1837 or VX-770 did not show any negative drug-drug interaction. CONCLUSION: N1303K has a severe gating defect, reduced ATP-dependence and aberrant response to ATP analogs. These results suggest a defective function of the NBDs in N1303K-CFTR. An improvement of channel function by GLPG1837 or VX-770 and an increase of Band C protein by VX-809 or VX-661 support a therapeutic strategy of combining CFTR potentiator and corrector for patients carrying the N1303K mutation.


Subject(s)
Chloride Channel Agonists/pharmacology , Cystic Fibrosis Transmembrane Conductance Regulator/agonists , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Mutant Proteins/drug effects , Adenosine Triphosphate/physiology , Aminophenols , Aminopyridines , Animals , Benzodioxoles , Blotting, Western , CHO Cells , Cricetinae , Cricetulus , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Indoles , Ion Channel Gating/drug effects , Mutation , Patch-Clamp Techniques , Phosphorylation , Quinolones , Structure-Activity Relationship
13.
J Med Chem ; 61(4): 1436-1449, 2018 02 22.
Article in English | MEDLINE | ID: mdl-29251932

ABSTRACT

Cystic fibrosis (CF) is a multiorgan disease of the lungs, sinuses, pancreas, and gastrointestinal tract that is caused by a dysfunction or deficiency of the cystic fibrosis transmembrane conductance regulator (CFTR) protein, an epithelial anion channel that regulates salt and water balance in the tissues in which it is expressed. To effectively treat the most prevalent patient population (F508del mutation), two biomolecular modulators are required: correctors to increase CFTR levels at the cell surface, and potentiators to allow the effective opening of the CFTR channel. Despite approved potentiator and potentiator/corrector combination therapies, there remains a high need to develop more potent and efficacious correctors. Herein, we disclose the discovery of a highly potent series of CFTR correctors and the structure-activity relationship (SAR) studies that guided the discovery of ABBV/GLPG-2222 (22), which is currently in clinical trials in patients harboring the F508del CFTR mutation on at least one allele.


Subject(s)
Benzoates/pharmacology , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis/drug therapy , Drug Discovery , Amides/chemical synthesis , Animals , Benzoates/chemical synthesis , Benzoates/pharmacokinetics , Chromans/chemical synthesis , Dogs , Humans , Mutant Proteins/drug effects , Rats , Structure-Activity Relationship
14.
J Med Chem ; 61(4): 1425-1435, 2018 02 22.
Article in English | MEDLINE | ID: mdl-29148763

ABSTRACT

Cystic fibrosis (CF) is caused by mutations in the gene for the cystic fibrosis transmembrane conductance regulator (CFTR). With the discovery of Ivacaftor and Orkambi, it has been shown that CFTR function can be partially restored by administering one or more small molecules. These molecules aim at either enhancing the amount of CFTR on the cell surface (correctors) or at improving the gating function of the CFTR channel (potentiators). Here we describe the discovery of a novel potentiator GLPG1837, which shows enhanced efficacy on CFTR mutants harboring class III mutations compared to Ivacaftor, the first marketed potentiator. The optimization of potency, efficacy, and pharmacokinetic profile will be described.


Subject(s)
Chloride Channel Agonists/chemistry , Cystic Fibrosis/drug therapy , Drug Discovery , Mutant Proteins/drug effects , Aminophenols/pharmacokinetics , Animals , Chloride Channel Agonists/pharmacokinetics , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Humans , Mutation , Pyrazoles/chemistry , Pyrazoles/pharmacokinetics , Quinolones/pharmacokinetics , Rats , Structure-Activity Relationship
15.
PLoS One ; 12(2): e0172832, 2017.
Article in English | MEDLINE | ID: mdl-28235006

ABSTRACT

Triple-negative breast cancer (TNBC) is an aggressive, highly recurrent breast cancer subtype, affecting approximately one-fifth of all breast cancer patients. Subpopulations of treatment-resistant cancer stem cells within the tumors are considered to contribute to disease recurrence. A potential druggable target for such cells is the maternal embryonic leucine-zipper kinase (MELK). MELK expression is upregulated in mammary stem cells and in undifferentiated cancers, where it correlates with poor prognosis and potentially mediates treatment resistance. Several MELK inhibitors have been developed, of which one, OTSSP167, is currently in clinical trials. In order to better understand how MELK and its inhibition influence TNBC, we verified its anti-proliferative and apoptotic effects in claudin-low TNBC cell lines MDA-MB-231 and SUM-159 using MTS assays and/or trypan blue viability assays together with analysis of PARP cleavage. Then, using microarrays, we explored which genes were affected by OTSSP167. We demonstrate that different sets of genes are regulated in MDA-MB-231 and SUM-159, but in both cell lines genes involved in cell cycle, mitosis and protein metabolism and folding were regulated. We identified p53 (TP53) as a potential upstream regulator of the regulated genes. Using western blot we found that OTSSP167 downregulates mutant p53 in all tested TNBC cell lines (MDA-MB-231, SUM-159, and BT-549), but upregulates wild-type p53 in the luminal A subtype MCF-7 cell line. We propose that OTSSP167 might have context-dependent or off-target effects, but that one consistent mechanism of action could involve the destabilization of mutant p53.


Subject(s)
Mutant Proteins/drug effects , Naphthyridines/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Triple Negative Breast Neoplasms/genetics , Tumor Suppressor Protein p53/drug effects , Apoptosis , Cell Cycle , Cell Line, Tumor , Cell Proliferation , DNA Mutational Analysis , Databases, Factual , Female , Humans , MCF-7 Cells , Mammary Glands, Human/cytology , Mitosis , Real-Time Polymerase Chain Reaction , Signal Transduction , Stem Cells/cytology , Triple Negative Breast Neoplasms/drug therapy , Up-Regulation
16.
Cancer ; 123(9): 1662-1673, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28055106

ABSTRACT

BACKGROUND: Nucleophosmin 1 (NPM1) is a nucleocytoplasmic shuttling protein mainly localized in the nucleolus. NPM1 is frequently mutated in acute myeloid leukemia (AML). NPM1c oligomerizes with wild-type nucleophosmin 1 (wt-NPM1), and this leads to its continuous cytoplasmic delocalization and contributes to leukemogenesis. Recent studies have shown that Cytoplasmic NPM1 (NPM1c) degradation leads to growth arrest and apoptosis of NPM1c AML cells and corrects wt-NPM1 normal nucleolar localization. METHODS: AML cells expressing wt-NPM1 or NPM1c or transfected with wt-NPM1 or NPM1c as well as wt-NPM1 and NPM1c AML xenograft mice were used. Cell growth was assessed with trypan blue or a CellTiter 96 proliferation kit. The cell cycle was studied with a propidium iodide (PI) assay. Caspase-mediated intrinsic apoptosis was assessed with annexin V/PI, the mitochondrial membrane potential, and poly(adenosine diphosphate ribose) polymerase cleavage. The expression of NPM1, p53, phosphorylated p53, and p21 was analyzed via immunoblotting. Localization was performed with confocal microscopy. The leukemia burden was evaluated by flow cytometry with an anti-human CD45 antibody. RESULTS: The imidazoquinoxaline 1-(3-methoxyphenyl)-N-methylimidazo[1,2-a]quinoxalin-4-amine (EAPB0503) induced selective proteasome-mediated degradation of NPM1c, restored wt-NPM1 nucleolar localization in NPM1c AML cells, and thus yielded selective growth arrest and apoptosis. Introducing NPM1c to cells normally harboring wt-NPM1 sensitized them to EAPB0503 and led to their growth arrest. Moreover, EAPB0503 selectively reduced the leukemia burden in NPM1c AML xenograft mice. CONCLUSIONS: These findings further reinforce the idea of targeting the NPM1c oncoprotein to eradicate leukemic cells and warrant a broader preclinical evaluation and then a clinical evaluation of this promising drug. Cancer 2017;123:1662-1673. © 2017 American Cancer Society.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Leukemia, Myeloid, Acute/drug therapy , Mutant Proteins/drug effects , Nuclear Proteins/drug effects , Quinoxalines/pharmacology , Animals , Annexin A5/drug effects , Annexin A5/metabolism , Cell Line, Tumor , Cell Nucleolus/metabolism , Cyclin-Dependent Kinase Inhibitor p21/drug effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cytoplasm/metabolism , Flow Cytometry , Humans , Immunoblotting , Leukemia, Myeloid, Acute/genetics , Mice , Microscopy, Confocal , Mutant Proteins/metabolism , Mutation , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nucleophosmin , Phosphorylation/drug effects , Poly(ADP-ribose) Polymerases/drug effects , Tumor Suppressor Protein p53/drug effects , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays
17.
Oncol Rep ; 37(1): 594-600, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27878254

ABSTRACT

Ganoderma lucidum polysaccharides (GLPs), isolated from spores, mycelia and fruiting bodies of Ganoderma lucidum, have been suggested to possess anticancer activities in a large number of basic studies. A recent survey revealed that GLP-induced inhibition of cancer cell growth was dependent on the existence of functional p53. However, the actual role of p53-mediated tumor-suppressing pathways in facilitating the anticancer effect of GLPs is still unclear. In the present study, we investigated the interaction between GLPs and mutant p53 that exists in more than half of the known types of cancers. Our results showed that GLPs reactivated mutant p53 in colorectal cancer HT29 (p53R273H) and SW480 (p53R273H&P309S) cells while applied alone or together with 5-fluorouracil (5-FU). This reactivation further induced cell growth inhibition and apoptosis. In addition, western blot assay and in vitro cell-free apoptosis assay suggested that the activation of mutant p53 was effective in both a transcriptional-dependent and -independent pathway. Altogether, our data demonstrated for the first time that GLPs show prominent anticancer activities by reactivating several types of mutant p53. Therefore, targeting mutant p53 by GLPs alongside other chemotherapeutics may be considered as a novel treatment strategy for cancer.


Subject(s)
Colorectal Neoplasms/genetics , Drug Resistance, Neoplasm/drug effects , Polysaccharides/pharmacology , Reishi/chemistry , Tumor Suppressor Protein p53/drug effects , Tumor Suppressor Protein p53/genetics , Apoptosis/drug effects , Cell Line, Tumor , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Drug Resistance, Neoplasm/genetics , Drug Synergism , Fluorouracil/pharmacology , Fluorouracil/therapeutic use , Genes, Tumor Suppressor/drug effects , Genes, Tumor Suppressor/physiology , HCT116 Cells , HT29 Cells , Humans , Mutant Proteins/drug effects , Mutant Proteins/physiology , Polysaccharides/isolation & purification
18.
Brain ; 139(11): 2891-2908, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27645800

ABSTRACT

No disease-modifying treatment exists for the fatal neurodegenerative polyglutamine disease known both as Machado-Joseph disease and spinocerebellar ataxia type 3. As a potential route to therapy, we identified small molecules that reduce levels of the mutant disease protein, ATXN3. Screens of a small molecule collection, including 1250 Food and Drug Administration-approved drugs, in a novel cell-based assay, followed by secondary screens in brain slice cultures from transgenic mice expressing the human disease gene, identified the atypical antipsychotic aripiprazole as one of the hits. Aripiprazole increased longevity in a Drosophila model of Machado-Joseph disease and effectively reduced aggregated ATXN3 species in flies and in brains of transgenic mice treated for 10 days. The aripiprazole-mediated decrease in ATXN3 abundance may reflect a complex response culminating in the modulation of specific components of cellular protein homeostasis. Aripiprazole represents a potentially promising therapeutic drug for Machado-Joseph disease and possibly other neurological proteinopathies.


Subject(s)
Antipsychotic Agents/therapeutic use , Aripiprazole/therapeutic use , Ataxin-3/metabolism , Machado-Joseph Disease/drug therapy , Machado-Joseph Disease/metabolism , Mutant Proteins/drug effects , Animals , Animals, Genetically Modified , Ataxin-3/genetics , Brain/drug effects , Brain/metabolism , Brain/ultrastructure , Disease Models, Animal , Drosophila , Drug Evaluation, Preclinical , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , HEK293 Cells/drug effects , HEK293 Cells/metabolism , HEK293 Cells/ultrastructure , Humans , Machado-Joseph Disease/genetics , Mice , Mutant Proteins/metabolism , Nerve Tissue Proteins/metabolism , Organ Culture Techniques , Peptides/genetics , Piperidines/pharmacology , Pyrans/pharmacology , Pyrazoles/pharmacology
19.
Nat Cell Biol ; 18(8): 897-909, 2016 08.
Article in English | MEDLINE | ID: mdl-27347849

ABSTRACT

In cancer, the tumour suppressor gene TP53 undergoes frequent missense mutations that endow mutant p53 proteins with oncogenic properties. Until now, a universal mutant p53 gain-of-function program has not been defined. By means of multi-omics: proteome, DNA interactome (chromatin immunoprecipitation followed by sequencing) and transcriptome (RNA sequencing/microarray) analyses, we identified the proteasome machinery as a common target of p53 missense mutants. The mutant p53-proteasome axis globally affects protein homeostasis, inhibiting multiple tumour-suppressive pathways, including the anti-oncogenic KSRP-microRNA pathway. In cancer cells, p53 missense mutants cooperate with Nrf2 (NFE2L2) to activate proteasome gene transcription, resulting in resistance to the proteasome inhibitor carfilzomib. Combining the mutant p53-inactivating agent APR-246 (PRIMA-1MET) with the proteasome inhibitor carfilzomib is effective in overcoming chemoresistance in triple-negative breast cancer cells, creating a therapeutic opportunity for treatment of solid tumours and metastasis with mutant p53.


Subject(s)
Mutant Proteins/drug effects , Mutation, Missense/drug effects , Proteasome Endopeptidase Complex/drug effects , Tumor Suppressor Protein p53/genetics , Animals , Antineoplastic Agents/pharmacology , Humans , Mice , MicroRNAs/genetics , Mutant Proteins/genetics , Mutation, Missense/genetics , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Neoplasm Metastasis/drug therapy , Neoplasm Metastasis/genetics , Oligopeptides/pharmacology , Proteasome Endopeptidase Complex/genetics , Proteome/drug effects , Quinuclidines/pharmacology , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Tumor Suppressor Protein p53/metabolism
20.
Int J Oncol ; 47(4): 1241-8, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26314369

ABSTRACT

TP53 mutation is a common event in many cancers, including thyroid carcinoma. Defective p53 activity promotes cancer resistance to therapies and a more malignant phenotype, acquiring oncogenic functions. Rescuing the function of mutant p53 (mutp53) protein is an attractive anticancer therapeutic strategy. Zn(II)-curc is a novel small molecule that has been shown to target mutp53 protein in several cancer cells, but its effect in thyroid cancer cells remains unclear. Here, we investigated whether Zn(II)-curc could affect p53 in thyroid cancer cells with both p53 mutation (R273H) and wild-type p53. Zn(II)-curc induced mutp53H273 downregulation and reactivation of wild-type functions, such as binding to canonical target promoters and target gene transactivation. This latter effect was similar to that induced by PRIMA-1. In addition, Zn(II)-curc triggered p53 target gene expression in wild-type p53-carrying cells. In combination treatments, Zn(II)-curc enhanced the antitumor activity of chemotherapeutic drugs, in both mutant and wild-type-carrying cancer cells. Taken together, our data indicate that Zn(II)-curc promotes the reactivation of p53 in thyroid cancer cells, providing in vitro evidence for a potential therapeutic approach in thyroid cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Curcumin/pharmacology , Mutant Proteins/drug effects , Thyroid Neoplasms/metabolism , Tumor Suppressor Protein p53/metabolism , Zinc Compounds/pharmacology , Blotting, Western , Cell Line, Tumor , Cell Survival/drug effects , Chromatin Immunoprecipitation , Humans , Mutant Proteins/genetics , Mutation , Reverse Transcriptase Polymerase Chain Reaction , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology , Tumor Suppressor Protein p53/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...