Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Proc Natl Acad Sci U S A ; 117(51): 32453-32463, 2020 12 22.
Article in English | MEDLINE | ID: mdl-33288711

ABSTRACT

Pathogenic mutations in the copper transporter ATP7B have been hypothesized to affect its protein interaction landscape contributing to loss of function and, thereby, to hepatic copper toxicosis in Wilson disease. Although targeting mutant interactomes was proposed as a therapeutic strategy, druggable interactors for rescue of ATP7B mutants remain elusive. Using proteomics, we found that the frequent H1069Q substitution promotes ATP7B interaction with HSP70, thus accelerating endoplasmic reticulum (ER) degradation of the mutant protein and consequent copper accumulation in hepatic cells. This prompted us to use an HSP70 inhibitor as bait in a bioinformatics search for structurally similar Food and Drug Administration-approved drugs. Among the hits, domperidone emerged as an effective corrector that recovered trafficking and function of ATP7B-H1069Q by impairing its exposure to the HSP70 proteostatic network. Our findings suggest that HSP70-mediated degradation can be safely targeted with domperidone to rescue ER-retained ATP7B mutants and, hence, to counter the onset of Wilson disease.


Subject(s)
Copper-Transporting ATPases/genetics , Copper-Transporting ATPases/metabolism , Domperidone/pharmacology , HSP70 Heat-Shock Proteins/metabolism , Hepatolenticular Degeneration/genetics , Benzimidazoles/chemistry , Benzimidazoles/pharmacology , Cells, Cultured , Copper/metabolism , Domperidone/chemistry , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , HSP70 Heat-Shock Proteins/antagonists & inhibitors , Hep G2 Cells , Hepatocytes/metabolism , Hepatolenticular Degeneration/drug therapy , Hepatolenticular Degeneration/metabolism , Hepatolenticular Degeneration/pathology , Humans , Mutation, Missense , Nipecotic Acids/chemistry , Nipecotic Acids/pharmacology , Protein Transport/drug effects , Protein Transport/genetics , Proteomics/methods
2.
Int J Mol Sci ; 22(21)2021 Nov 04.
Article in English | MEDLINE | ID: mdl-34769402

ABSTRACT

Cystic fibrosis (CF) is caused by loss of function of the CFTR chloride channel. A substantial number of CF patients carry nonsense mutations in the CFTR gene. These patients cannot directly benefit from pharmacological correctors and potentiators that have been developed for other types of CFTR mutations. We evaluated the efficacy of combinations of drugs targeting at various levels the effects of nonsense mutations: SMG1i to protect CFTR mRNA from nonsense-mediated decay (NMD), G418 and ELX-02 for readthrough, VX-809 and VX-445 to promote protein maturation and function, PTI-428 to enhance CFTR protein synthesis. We found that the extent of rescue and sensitivity to the various agents is largely dependent on the type of mutation, with W1282X and R553X being the mutations most and least sensitive to pharmacological treatments, respectively. In particular, W1282X-CFTR was highly responsive to NMD suppression by SMG1i but also required treatment with VX-445 corrector to show function. In contrast, G542X-CFTR required treatment with readthrough agents and VX-809. Importantly, we never found cooperativity between the NMD inhibitor and readthrough compounds. Our results indicate that treatment of CF patients with nonsense mutations requires a precision medicine approach with the design of specific drug combinations for each mutation.


Subject(s)
Aminopyridines/pharmacology , Benzodioxoles/pharmacology , Codon, Nonsense , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis/drug therapy , Nonsense Mediated mRNA Decay/drug effects , Pyrazoles/pharmacology , Pyridines/pharmacology , Pyrrolidines/pharmacology , Bronchi/drug effects , Chloride Channel Agonists/pharmacology , Cystic Fibrosis/genetics , Cystic Fibrosis/pathology , Epithelial Cells/drug effects , Humans
3.
Int J Mol Sci ; 22(10)2021 May 17.
Article in English | MEDLINE | ID: mdl-34067708

ABSTRACT

Deletion of phenylalanine at position 508 (F508del) in the CFTR chloride channel is the most frequent mutation in cystic fibrosis (CF) patients. F508del impairs the stability and folding of the CFTR protein, thus resulting in mistrafficking and premature degradation. F508del-CFTR defects can be overcome with small molecules termed correctors. We investigated the efficacy and properties of VX-445, a newly developed corrector, which is one of the three active principles present in a drug (Trikafta®/Kaftrio®) recently approved for the treatment of CF patients with F508del mutation. We found that VX-445, particularly in combination with type I (VX-809, VX-661) and type II (corr-4a) correctors, elicits a large rescue of F508del-CFTR function. In particular, in primary bronchial epithelial cells of CF patients, the maximal rescue obtained with corrector combinations including VX-445 was close to 60-70% of CFTR function in non-CF cells. Despite this high efficacy, analysis of ubiquitylation, resistance to thermoaggregation, protein half-life, and subcellular localization revealed that corrector combinations did not fully normalize F508del-CFTR behavior. Our study indicates that it is still possible to further improve mutant CFTR rescue with the development of corrector combinations having maximal effects on mutant CFTR structural and functional properties.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/drug effects , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Pyrazoles/pharmacology , Pyridines/pharmacology , Pyrrolidines/pharmacology , Aminophenols/pharmacology , Aminopyridines/pharmacology , Benzodioxoles/pharmacology , Bronchi/drug effects , Bronchi/metabolism , Chloride Channels/genetics , Chloride Channels/metabolism , Cystic Fibrosis/drug therapy , Cystic Fibrosis/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Drug Combinations , Epithelial Cells/metabolism , Humans , Indoles/pharmacology , Protein Folding/drug effects , Pyrazoles/metabolism , Pyridines/metabolism , Pyrrolidines/metabolism , Quinolines/pharmacology
4.
Molecules ; 26(5)2021 Feb 26.
Article in English | MEDLINE | ID: mdl-33652850

ABSTRACT

Cystic fibrosis (CF) is a genetic disease caused by mutations that impair the function of the CFTR chloride channel. The most frequent mutation, F508del, causes misfolding and premature degradation of CFTR protein. This defect can be overcome with pharmacological agents named "correctors". So far, at least three different classes of correctors have been identified based on the additive/synergistic effects that are obtained when compounds of different classes are combined together. The development of class 2 correctors has lagged behind that of compounds belonging to the other classes. It was shown that the efficacy of the prototypical class 2 corrector, the bithiazole corr-4a, could be improved by generating conformationally-locked bithiazoles. In the present study, we investigated the effect of tricyclic pyrrolothiazoles as analogues of constrained bithiazoles. Thirty-five compounds were tested using the functional assay based on the halide-sensitive yellow fluorescent protein (HS-YFP) that measured CFTR activity. One compound, having a six atom carbocyle central ring in the tricyclic pyrrolothiazole system and bearing a pivalamide group at the thiazole moiety and a 5-chloro-2-methoxyphenyl carboxamide at the pyrrole ring, significantly increased F508del-CFTR activity. This compound could lead to the synthesis of a novel class of CFTR correctors.


Subject(s)
Benzodioxoles/pharmacology , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis/drug therapy , Mutant Proteins/genetics , Aminoimidazole Carboxamide/chemistry , Benzodioxoles/chemistry , Cystic Fibrosis/genetics , Cystic Fibrosis/pathology , Cystic Fibrosis Transmembrane Conductance Regulator/drug effects , Humans , Mutation/drug effects , Mutation/genetics , Protein Folding/drug effects , Thiazoles/chemistry
5.
ACS Pharmacol Transl Sci ; 7(6): 1807-1822, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38898954

ABSTRACT

Over the past few years, l-iminosugars have revealed attractive pharmacological properties for managing rare diseases including Cystic Fibrosis (CF). The iminosugar N-butyl-l-deoxynojirimycin (l-NBDNJ, ent-1), prepared by a carbohydrate-based route, was herein evaluated for its anti-inflammatory and anti-infective potential in models of CF lung disease infection. A significant decrease in the bacterial load in the airways was observed in the murine model of Pseudomonas aeruginosa chronic infection in the presence of l-NBDNJ, also accompanied by a modest reduction of inflammatory cells. Mechanistic insights into the observed activity revealed that l-NBDNJ interferes with the expression of proteins regulating cytoskeleton assembly and organization of the host cell, downregulates the main virulence factors of P. aeruginosa involved in the host response, and affects pathogen adhesion to human cells. These findings along with the observation of the absence of an in vitro bacteriostatic/bactericidal action of l-NBDNJ suggest the potential use of this glycomimetic as an antivirulence agent in the management of CF lung disease.

6.
Sci Rep ; 13(1): 7604, 2023 05 10.
Article in English | MEDLINE | ID: mdl-37165082

ABSTRACT

F508del, the most frequent mutation in cystic fibrosis (CF), impairs the stability and folding of the CFTR chloride channel, thus resulting in intracellular retention and CFTR degradation. The F508del defect can be targeted with pharmacological correctors, such as VX-809 and VX-445, that stabilize CFTR and improve its trafficking to plasma membrane. Using a functional test to evaluate a panel of chemical compounds, we have identified tricyclic pyrrolo-quinolines as novel F508del correctors with high efficacy on primary airway epithelial cells from CF patients. The most effective compound, PP028, showed synergy when combined with VX-809 and VX-661 but not with VX-445. By testing the ability of correctors to stabilize CFTR fragments of different length, we found that VX-809 is effective on the amino-terminal portion of the protein that includes the first membrane-spanning domain (amino acids 1-387). Instead, PP028 and VX-445 only show a stabilizing effect when the second membrane-spanning domain is included (amino acids 1-1181). Our results indicate that tricyclic pyrrolo-quinolines are a novel class of CFTR correctors that, similarly to VX-445, interact with CFTR at a site different from that of VX-809. Tricyclic pirrolo-quinolines may represent novel CFTR correctors suitable for combinatorial pharmacological treatments to treat the basic defect in CF.


Subject(s)
Cystic Fibrosis , Quinolines , Humans , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Chloride Channels/genetics , Quinolines/therapeutic use , Cystic Fibrosis/drug therapy , Cystic Fibrosis/genetics , Cystic Fibrosis/metabolism , Benzodioxoles/pharmacology , Benzodioxoles/therapeutic use , Aminopyridines/pharmacology , Aminopyridines/therapeutic use , Mutation
7.
Proc Natl Acad Sci U S A ; 106(17): 6933-8, 2009 Apr 28.
Article in English | MEDLINE | ID: mdl-19369210

ABSTRACT

The recent characterization of the prokaryotic Cys(2)His(2) zinc-finger domain, identified in Ros protein from Agrobacterium tumefaciens, has demonstrated that, although possessing a similar zinc coordination sphere, this domain is structurally very different from its eukaryotic counterpart. A search in the databases has identified approximately 300 homologues with a high sequence identity to the Ros protein, including the amino acids that form the extensive hydrophobic core in Ros. Surprisingly, the Cys(2)His(2) zinc coordination sphere is generally poorly conserved in the Ros homologues, raising the question of whether the zinc ion is always preserved in these proteins. Here, we present a functional and structural study of a point mutant of Ros protein, Ros(56-142)C82D, in which the second coordinating cysteine is replaced by an aspartate, 5 previously-uncharacterized representative Ros homologues from Mesorhizobium loti, and 2 mutants of the homologues. Our results indicate that the prokaryotic zinc-finger domain, which in Ros protein tetrahedrally coordinates Zn(II) through the typical Cys(2)His(2) coordination, in Ros homologues can either exploit a CysAspHis(2) coordination sphere, previously never described in DNA binding zinc finger domains to our knowledge, or lose the metal, while still preserving the DNA-binding activity. We demonstrate that this class of prokaryotic zinc-finger domains is structurally very adaptable, and surprisingly single mutations can transform a zinc-binding domain into a nonzinc-binding domain and vice versa, without affecting the DNA-binding ability. In light of our findings an evolutionary link between the prokaryotic and eukaryotic zinc-finger domains, based on bacteria-to-eukaryota horizontal gene transfer, is discussed.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Zinc Fingers , Zinc/chemistry , Zinc/metabolism , Alphaproteobacteria/chemistry , Alphaproteobacteria/genetics , Alphaproteobacteria/metabolism , Amino Acid Sequence , Bacterial Proteins/genetics , Cations , Hydrophobic and Hydrophilic Interactions , Mass Spectrometry , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Protein Structure, Secondary , Sequence Alignment , Sequence Homology, Amino Acid
8.
Front Pharmacol ; 9: 1464, 2018.
Article in English | MEDLINE | ID: mdl-30618756

ABSTRACT

The mutation F508del, responsible for a majority of cystic fibrosis cases, provokes the instability and misfolding of the CFTR chloride channel. Pharmacological recovery of F508del-CFTR may be obtained with small molecules called correctors. However, treatment with a single corrector in vivo and in vitro only leads to a partial rescue, a consequence of cell quality control systems that still detect F508del-CFTR as a defective protein causing its degradation. We tested the effect of spautin-1 on F508del-CFTR since it is an inhibitor of USP10 deubiquitinase and of autophagy, a target and a biological process that have been associated with cystic fibrosis and mutant CFTR. We found that short-term treatment of cells with spautin-1 downregulates the function and expression of F508del-CFTR despite the presence of corrector VX-809, a finding obtained in multiple cell models and assays. In contrast, spautin-1 was ineffective on wild type CFTR. Silencing and upregulation of USP13 (another target of spautin-1) but not of USP10, had opposite effects on F508del-CFTR expression/function. In contrast, modulation of autophagy with known activators or inhibitors did not affect F508del-CFTR. Our results identify spautin-1 as a novel chemical probe to investigate the molecular mechanisms that prevent full rescue of mutant CFTR.

9.
Sci Rep ; 7(1): 1224, 2017 04 27.
Article in English | MEDLINE | ID: mdl-28450740

ABSTRACT

Protein synthesis is traditionally associated with specific cytoplasmic compartments. We now show that OFD1, a centrosomal/basal body protein, interacts with components of the Preinitiation complex of translation (PIC) and of the eukaryotic Initiation Factor (eIF)4F complex and modulates the translation of specific mRNA targets in the kidney. We demonstrate that OFD1 cooperates with the mRNA binding protein Bicc1 to functionally control the protein synthesis machinery at the centrosome where also the PIC and eIF4F components were shown to localize in mammalian cells. Interestingly, Ofd1 and Bicc1 are both involved in renal cystogenesis and selected targets were shown to accumulate in two models of inherited renal cystic disease. Our results suggest a possible role for the centrosome as a specialized station to modulate translation for specific functions of the nearby ciliary structures and may provide functional clues for the understanding of renal cystic disease.


Subject(s)
Centrosome/metabolism , Gene Expression Regulation , Protein Biosynthesis , Protein Interaction Mapping , Proteins/metabolism , RNA-Binding Proteins/metabolism , HEK293 Cells , HeLa Cells , Humans
10.
JCI Insight ; 2(24)2017 12 21.
Article in English | MEDLINE | ID: mdl-29263295

ABSTRACT

The genome-wide activity of transcription factors (TFs) on multiple regulatory elements precludes their use as gene-specific regulators. Here we show that ectopic expression of a TF in a cell-specific context can be used to silence the expression of a specific gene as a therapeutic approach to regulate gene expression in human disease. We selected the TF Krüppel-like factor 15 (KLF15) based on its putative ability to recognize a specific DNA sequence motif present in the rhodopsin (RHO) promoter and its lack of expression in terminally differentiated rod photoreceptors (the RHO-expressing cells). Adeno-associated virus (AAV) vector-mediated ectopic expression of KLF15 in rod photoreceptors of pigs enables Rho silencing with limited genome-wide transcriptional perturbations. Suppression of a RHO mutant allele by KLF15 corrects the phenotype of a mouse model of retinitis pigmentosa with no observed toxicity. Cell-specific-context conditioning of TF activity may prove a novel mode for somatic gene-targeted manipulation.


Subject(s)
Gene Silencing , Gene Targeting/methods , Kruppel-Like Transcription Factors/genetics , Nuclear Proteins/genetics , Rhodopsin/genetics , Animals , Dependovirus/genetics , Ectopic Gene Expression , Female , Genetic Therapy/methods , Genetic Vectors , Kruppel-Like Transcription Factors/physiology , Mice, Transgenic , Mutation , Nuclear Proteins/physiology , Retinal Rod Photoreceptor Cells/metabolism , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/therapy , Rhodopsin/metabolism , Swine
11.
Elife ; 5: e12242, 2016 Mar 14.
Article in English | MEDLINE | ID: mdl-26974343

ABSTRACT

Transcription factors (TFs) operate by the combined activity of their DNA-binding domains (DBDs) and effector domains (EDs) enabling the coordination of gene expression on a genomic scale. Here we show that in vivo delivery of an engineered DNA-binding protein uncoupled from the repressor domain can produce efficient and gene-specific transcriptional silencing. To interfere with RHODOPSIN (RHO) gain-of-function mutations we engineered the ZF6-DNA-binding protein (ZF6-DB) that targets 20 base pairs (bp) of a RHOcis-regulatory element (CRE) and demonstrate Rho specific transcriptional silencing upon adeno-associated viral (AAV) vector-mediated expression in photoreceptors. The data show that the 20 bp-long genomic DNA sequence is necessary for RHO expression and that photoreceptor delivery of the corresponding cognate synthetic trans-acting factor ZF6-DB without the intrinsic transcriptional repression properties of the canonical ED blocks Rho expression with negligible genome-wide transcript perturbations. The data support DNA-binding-mediated silencing as a novel mode to treat gain-of-function mutations.


Subject(s)
DNA-Binding Proteins/metabolism , DNA/metabolism , Gene Silencing , Recombinant Proteins/metabolism , Rhodopsin/biosynthesis , Transcription, Genetic , Adenoviridae/genetics , DNA-Binding Proteins/genetics , Gene Expression , Genetic Vectors , Protein Binding , Recombinant Proteins/genetics , Transduction, Genetic
12.
Microbiol Res ; 168(2): 65-72, 2013 Feb 22.
Article in English | MEDLINE | ID: mdl-23103380

ABSTRACT

The enolase EnoA1 of Lactobacillus plantarum is here shown to interact with human plasminogen (Plg). By sequence alignment of EnoA1 with Streptococcus pneumoniae and Bifidobacterium lactis enolases, we identified BS1 and BS2 Plg-binding sites. A structure prediction of EnoA1 showed lysine residues in position 255 (BS2), and 422 (BS1) exposed on protein surface. A lysine residue in position 259 was as well identified as surface-exposed amino acid. The enoA1 gene was site directed-mutagenized to generate four mutated proteins, carrying K255A, K259A, K422A and K259A/K422A substitutions. The functional role of these lysine residues was assessed evaluating specific Plg-binding activity of the mutated proteins. While the binding activity of the mutated proteins was drastically reduced, the residual enzymatic activity was more than 50% of EnoA1. Our results show that L. plantarum EnoA1 exhibits the Plg-BS1, and the Plg-BS2 extending up to the lysine residue in position 259, therefore consisting of 12-aa residues instead of 9-aa residues described in S. pneumoniae. A test performed on whole cells of L. plantarum, demonstrated that after inducing conversion of the cell-bound plasminogen to plasmin, this was released into the medium, unlike the mechanism reported for most pathogens, that retained plasmin bound to the cell surface.


Subject(s)
Binding Sites/genetics , Lactobacillus plantarum/enzymology , Phosphopyruvate Hydratase/metabolism , Plasminogen/metabolism , Amino Acid Sequence , Fibrinolysin/metabolism , Humans , Lactobacillus plantarum/genetics , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Phosphopyruvate Hydratase/chemistry , Phosphopyruvate Hydratase/genetics , Phosphopyruvate Hydratase/isolation & purification , Probiotics , Protein Binding , Sequence Alignment , Sequence Analysis, DNA
13.
Biopolymers ; 93(5): 434-41, 2010 May.
Article in English | MEDLINE | ID: mdl-19938072

ABSTRACT

The article describes the use of a PNA duplex (PNA zipper) as a tool to dimerize or bring in close proximity two polypeptides or protein domains. The amino acid sequence to be dimerized is covalently bound to complementary PNA sequences. Annealing of the PNA strands results in dimer formation. To test the ability of the "PNA-zipper" as a dimerization tool, we designed a GCN4 mimetic, where the leucine-zipper dimerization domain was replaced by the PNA zipper, whereas the basic DNA-binding domain was covalently attached to the PNA. The molecule was assembled by chemical ligation of the peptide corresponding to the DNA-binding domain of GCN4 modified with a succinyl thioester with two complementary PNAs harboring a cysteine residue. Electromobility-shift experiments show the ability of the PNA zipper-GCN4 to bind selected DNA duplexes. The PNA zipper-GCN4 binds both the TRE and CRE DNA sites, but it does not bind TRE and CRE mutants containing even a single base mutation, as the native GCN4. The ability to fold upon complexation with DNA was investigated by CD. A good correlation between the ability of the PNA zipper-GCN4 to fold into alpha helices and the ability to bind DNA was found.


Subject(s)
Basic-Leucine Zipper Transcription Factors/chemistry , Biomimetics , Peptide Nucleic Acids/chemistry , Amino Acid Sequence , Circular Dichroism , Computer Simulation , Dimerization , Models, Molecular , Molecular Sequence Data , Molecular Structure , Protein Folding
14.
J Biol Chem ; 282(46): 33336-33345, 2007 Nov 16.
Article in English | MEDLINE | ID: mdl-17827499

ABSTRACT

The DNA-binding protein CTCF (CCCTC binding factor) mediates enhancer blocking insulation at sites throughout the genome and plays an important role in regulating allele-specific expression at the Igf2/H19 locus and at other imprinted loci. Evidence is also accumulating that CTCF is involved in large scale organization of genomic chromatin. Although CTCF has 11 zinc fingers, we show here that only 4 of these are essential to strong binding and that they recognize a core 12-bp DNA sequence common to most CTCF sites. By deleting individual fingers and mutating individual sites, we determined the orientation of binding. Furthermore, we were able to identify the specific finger and its point of DNA interaction that are responsible for the loss of CTCF binding when CpG residues are methylated in the imprinted Igf2/H19 locus. This single interaction appears to be critical for allele-specific binding and insulation by CTCF.


Subject(s)
DNA-Binding Proteins/physiology , DNA/chemistry , Genomic Imprinting , Repressor Proteins/physiology , Base Sequence , Binding Sites , CCCTC-Binding Factor , CpG Islands , DNA Methylation , DNA-Binding Proteins/chemistry , Enhancer Elements, Genetic , Humans , Molecular Sequence Data , Protein Binding , Protein Structure, Tertiary , Repressor Proteins/chemistry , Transcription Factors/metabolism , Zinc Fingers
SELECTION OF CITATIONS
SEARCH DETAIL