Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Am J Hum Genet ; 110(3): 531-547, 2023 03 02.
Article in English | MEDLINE | ID: mdl-36809767

ABSTRACT

Familial dysautonomia (FD) is a rare neurodegenerative disease caused by a splicing mutation in elongator acetyltransferase complex subunit 1 (ELP1). This mutation leads to the skipping of exon 20 and a tissue-specific reduction of ELP1, mainly in the central and peripheral nervous systems. FD is a complex neurological disorder accompanied by severe gait ataxia and retinal degeneration. There is currently no effective treatment to restore ELP1 production in individuals with FD, and the disease is ultimately fatal. After identifying kinetin as a small molecule able to correct the ELP1 splicing defect, we worked on its optimization to generate novel splicing modulator compounds (SMCs) that can be used in individuals with FD. Here, we optimize the potency, efficacy, and bio-distribution of second-generation kinetin derivatives to develop an oral treatment for FD that can efficiently pass the blood-brain barrier and correct the ELP1 splicing defect in the nervous system. We demonstrate that the novel compound PTC258 efficiently restores correct ELP1 splicing in mouse tissues, including brain, and most importantly, prevents the progressive neuronal degeneration that is characteristic of FD. Postnatal oral administration of PTC258 to the phenotypic mouse model TgFD9;Elp1Δ20/flox increases full-length ELP1 transcript in a dose-dependent manner and leads to a 2-fold increase in functional ELP1 in the brain. Remarkably, PTC258 treatment improves survival, gait ataxia, and retinal degeneration in the phenotypic FD mice. Our findings highlight the great therapeutic potential of this novel class of small molecules as an oral treatment for FD.


Subject(s)
Dysautonomia, Familial , Neurodegenerative Diseases , Retinal Degeneration , Mice , Animals , Dysautonomia, Familial/genetics , Kinetin , Gait Ataxia , Administration, Oral
2.
Elife ; 112022 02 09.
Article in English | MEDLINE | ID: mdl-35137690

ABSTRACT

Antibiotic-resistant Neisseria gonorrhoeae (Ng) are an emerging public health threat due to increasing numbers of multidrug resistant (MDR) organisms. We identified two novel orally active inhibitors, PTC-847 and PTC-672, that exhibit a narrow spectrum of activity against Ng including MDR isolates. By selecting organisms resistant to the novel inhibitors and sequencing their genomes, we identified a new therapeutic target, the class Ia ribonucleotide reductase (RNR). Resistance mutations in Ng map to the N-terminal cone domain of the α subunit, which we show here is involved in forming an inhibited α4ß4 state in the presence of the ß subunit and allosteric effector dATP. Enzyme assays confirm that PTC-847 and PTC-672 inhibit Ng RNR and reveal that allosteric effector dATP potentiates the inhibitory effect. Oral administration of PTC-672 reduces Ng infection in a mouse model and may have therapeutic potential for treatment of Ng that is resistant to current drugs.


Subject(s)
Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial/drug effects , Gonorrhea/drug therapy , Pyridines/pharmacology , Ribonucleotide Reductases/metabolism , Allosteric Regulation , Animals , Deoxyadenine Nucleotides/metabolism , Disease Models, Animal , Escherichia coli/drug effects , Female , Gonorrhea/metabolism , Humans , Mice , Mice, Inbred BALB C , Microbial Sensitivity Tests/methods , Neisseria gonorrhoeae/drug effects
3.
Antimicrob Agents Chemother ; 60(12): 7060-7066, 2016 12.
Article in English | MEDLINE | ID: mdl-27620477

ABSTRACT

PTC725 is a small molecule NS4B-targeting inhibitor of hepatitis C virus (HCV) genotype (gt) 1 RNA replication that lacks activity against HCV gt2. We analyzed the Los Alamos HCV sequence database to predict susceptible/resistant HCV gt's according to the prevalence of known resistance-conferring amino acids in the NS4B protein. Our analysis predicted that HCV gt3 would be highly susceptible to the activity of PTC725. Indeed, PTC725 was shown to be active against a gt3 subgenomic replicon with a 50% effective concentration of ∼5 nM. De novo resistance selection identified mutations encoding amino acid substitutions mapping to the first predicted transmembrane region of NS4B, a finding consistent with results for PTC725 and other NS4B-targeting compounds against HCV gt1. This is the first report of the activity of an NS4B targeting compound against HCV gt3. In addition, we have identified previously unreported amino acid substitutions selected by PTC725 treatment which further demonstrate that these compounds target the NS4B first transmembrane region.


Subject(s)
Antiviral Agents/pharmacology , Hepacivirus/drug effects , Hepacivirus/genetics , Indoles/pharmacology , Sulfonamides/pharmacology , Viral Nonstructural Proteins/genetics , Amino Acid Substitution , Cell Line, Tumor , Drug Resistance, Viral/drug effects , Drug Resistance, Viral/genetics , Genome, Viral , Genotype , Humans , Mutation , Replicon/drug effects , Viral Nonstructural Proteins/metabolism
4.
Bioorg Med Chem Lett ; 26(2): 594-601, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26652483

ABSTRACT

A novel series of 2-(4-sulfonamidophenyl)-indole 3-carboxamides was identified and optimized for activity against the HCV genotype 1b replicon resulting in compounds with potent and selective activity. Further evaluation of this series demonstrated potent activity across HCV genotypes 1a, 2a and 3a. Compound 4z had reduced activity against HCV genotype 1b replicons containing single mutations in the NS4B coding sequence (F98C and V105M) indicating that NS4B is the target. This novel series of 2-(4-sulfonamidophenyl)-indole 3-carboxamides serves as a promising starting point for a pan-genotype HCV discovery program.


Subject(s)
Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Hepacivirus/drug effects , Indoles/chemistry , Indoles/pharmacology , Viral Nonstructural Proteins/metabolism , Amino Acid Sequence , Hepacivirus/chemistry , Hepacivirus/genetics , Hepacivirus/metabolism , Hepatitis C/drug therapy , Humans , Molecular Sequence Data , Mutation , Replicon/drug effects , Sulfonamides/chemistry , Sulfonamides/pharmacology , Viral Nonstructural Proteins/antagonists & inhibitors , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/genetics
5.
Bioorg Med Chem Lett ; 25(4): 781-6, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25613678

ABSTRACT

A structure-activity relationship investigation of various 6-(azaindol-2-yl)pyridine-3-sulfonamides using the HCV replicon cell culture assay led to the identification of a potent series of 7-azaindoles that target the hepatitis C virus NS4B. Compound 2ac, identified via further optimization of the series, has excellent potency against the HCV 1b replicon with an EC50 of 2nM and a selectivity index of >5000 with respect to cellular GAPDH RNA. Compound 2ac also has excellent oral plasma exposure levels in rats, dogs and monkeys and has a favorable liver to plasma distribution profile in rats.


Subject(s)
Hepacivirus/enzymology , Pyridines/chemistry , Pyridines/pharmacology , Sulfonamides/pharmacology , Viral Nonstructural Proteins/antagonists & inhibitors , Animals , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Dogs , Hepacivirus/drug effects , Humans , Macaca fascicularis , Rats , Structure-Activity Relationship
6.
J Med Chem ; 57(5): 2121-35, 2014 Mar 13.
Article in English | MEDLINE | ID: mdl-24266880

ABSTRACT

A novel, potent, and orally bioavailable inhibitor of hepatitis C RNA replication targeting NS4B, compound 4t (PTC725), has been identified through chemical optimization of the 6-(indol-2-yl)pyridine-3-sulfonamide 2 to improve DMPK and safety properties. The focus of the SAR investigations has been to identify the optimal combination of substituents at the indole N-1, C-5, and C-6 positions and the sulfonamide group to limit the potential for in vivo oxidative metabolism and to achieve an acceptable pharmacokinetic profile. Compound 4t has excellent potency against the HCV 1b replicon, with an EC50 = 2 nM and a selectivity index of >5000 with respect to cellular GAPDH. Compound 4t has an overall favorable pharmacokinetic profile with oral bioavailability values of 62%, 78%, and 18% in rats, dogs, and monkeys, respectively, as well as favorable tissue distribution properties with a liver to plasma exposure ratio of 25 in rats.


Subject(s)
Antiviral Agents/pharmacology , Hepacivirus/drug effects , Sulfonamides/pharmacology , Administration, Oral , Animals , Antiviral Agents/administration & dosage , Antiviral Agents/pharmacokinetics , Area Under Curve , Biological Availability , Dogs , Haplorhini , Humans , Rats , Structure-Activity Relationship , Sulfonamides/administration & dosage , Sulfonamides/pharmacokinetics
7.
Antimicrob Agents Chemother ; 57(7): 3250-61, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23629699

ABSTRACT

While new direct-acting antiviral agents for the treatment of chronic hepatitis C virus (HCV) infection have been approved, there is a continued need for novel antiviral agents that act on new targets and can be used in combination with current therapies to enhance efficacy and to restrict the emergence of drug-resistant viral variants. To this end, we have identified a novel class of small molecules, exemplified by PTC725, that target the nonstructural protein 4B (NS4B). PTC725 inhibited HCV 1b (Con1) replicons with a 50% effective concentration (EC50) of 1.7 nM and an EC90 of 9.6 nM and demonstrated a >1,000-fold selectivity window with respect to cytotoxicity. The compounds were fully active against HCV replicon mutants that are resistant to inhibitors of NS3 protease and NS5B polymerase. Replicons selected for resistance to PTC725 harbored amino acid substitutions F98L/C and V105M in NS4B. Anti-replicon activity of PTC725 was additive to synergistic in combination with alpha interferon or with inhibitors of HCV protease and polymerase. Immunofluorescence microscopy demonstrated that neither the HCV inhibitors nor the F98C substitution altered the subcellular localization of NS4B or NS5A in replicon cells. Oral dosing of PTC725 showed a favorable pharmacokinetic profile with high liver and plasma exposure in mice and rats. Modeling of dosing regimens in humans indicates that a once-per-day or twice-per-day oral dosing regimen is feasible. Overall, the preclinical data support the development of PTC725 for use in the treatment of chronic HCV infection.


Subject(s)
Antiviral Agents/metabolism , Antiviral Agents/pharmacology , Hepacivirus/drug effects , Hepatitis C/drug therapy , Indoles/pharmacology , Sulfonamides/pharmacology , Viral Nonstructural Proteins/metabolism , Amino Acid Substitution , Animals , Antiviral Agents/pharmacokinetics , Cell Line, Tumor , Drug Resistance, Viral/genetics , Drug Synergism , Humans , Indoles/metabolism , Indoles/pharmacokinetics , Interferon-alpha/pharmacology , Male , Mice , Microbial Sensitivity Tests , Rats , Rats, Sprague-Dawley , Sulfonamides/metabolism , Sulfonamides/pharmacokinetics , Viral Nonstructural Proteins/genetics , Virus Replication/drug effects
8.
Nucleic Acids Res ; 36(7): 2338-52, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18299285

ABSTRACT

U1 interference (U1i) is a novel method to block gene expression. U1i requires expression of a 5'-end-mutated U1 snRNA designed to base pair to the 3'-terminal exon of the target gene's pre-mRNA that leads to inhibition of polyadenylation. Here, we show U1i is robust (> or =95%) and a 10-nt target length is sufficient for good silencing. Surprisingly, longer U1 snRNAs, which could increase annealing to the target, fail to improve silencing. Extensive mutagenesis of the 10-bp U1 snRNA:target duplex shows that any single mismatch different from GU at positions 3-8, destroys silencing. However, mismatches within the other positions give partial silencing, suggesting that off-target inhibition could occur. The specificity of U1i may be enhanced, however, by the fact that silencing is impaired by RNA secondary structure or by splicing factors binding nearby, the latter mediated by Arginine-Serine (RS) domains. U1i inhibition can be reconstituted in vivo by tethering of RS domains of U1-70K and U2AF65. These results help to: (i) define good target sites for U1i; (ii) identify and understand natural cellular examples of U1i; (iii) clarify the contribution of hydrogen bonding to U1i and to U1 snRNP binding to 5' splice sites and (iv) understand the mechanism of U1i.


Subject(s)
RNA Interference , RNA Precursors/chemistry , RNA, Messenger/chemistry , RNA, Small Nuclear/chemistry , Base Pair Mismatch , Binding Sites , HeLa Cells , Humans , Hydrogen Bonding , Nuclear Proteins/chemistry , Nucleic Acid Conformation , Point Mutation , Protein Structure, Tertiary , RNA Precursors/metabolism , RNA Splicing , RNA, Messenger/metabolism , RNA, Small Nuclear/metabolism , RNA-Binding Proteins/chemistry , Regulatory Sequences, Ribonucleic Acid , Serine-Arginine Splicing Factors
9.
Proc Natl Acad Sci U S A ; 100(14): 8264-9, 2003 Jul 08.
Article in English | MEDLINE | ID: mdl-12826613

ABSTRACT

Reducing or eliminating expression of a given gene is likely to require multiple methods to ensure coverage of all of the genes in a given mammalian cell. We and others [Furth, P. A., Choe, W. T., Rex, J. H., Byrne, J. C., and Baker, C. C. (1994) Mol. Cell. Biol. 14, 5278-5289] have previously shown that U1 small nuclear (sn) RNA, both natural or with 5' end mutations, can specifically inhibit reporter gene expression in mammalian cells. This inhibition occurs when the U1 snRNA 5' end base pairs near the polyadenylation signal of the reporter gene's pre-mRNA. This base pairing inhibits poly(A) tail addition, a key, nearly universal step in mRNA biosynthesis, resulting in degradation of the mRNA. Here we demonstrate that expression of endogenous mammalian genes can be efficiently inhibited by transiently or stably expressed 5' end-mutated U1 snRNA. Also, we determine the inhibitory mechanism and establish a set of rules to use this technique and to improve the efficiency of inhibition. Two U1 snRNAs base paired to a single pre-mRNA act synergistically, resulting in up to 700-fold inhibition of the expression of specific reporter genes and 25-fold inhibition of endogenous genes. Surprisingly, distance from the U1 snRNA binding site to the poly(A) signal is not critical for inhibition, instead the U1 snRNA must be targeted to the terminal exon of the pre-mRNA. This could reflect a disruption by the 5' end-mutated U1 snRNA of the definition of the terminal exon as described by the exon definition model.


Subject(s)
RNA Precursors/antagonists & inhibitors , RNA, Small Nuclear/pharmacology , 3' Untranslated Regions/genetics , 5' Untranslated Regions/genetics , Base Pairing , Binding Sites , Drug Synergism , Enhancer Elements, Genetic , Fibroblasts/cytology , Gene Expression Regulation/drug effects , Genes, Reporter , HeLa Cells , Humans , Introns/genetics , Luciferases/genetics , Nucleic Acid Conformation , Nucleic Acid Hybridization , Poly A/metabolism , Promoter Regions, Genetic , RNA Precursors/genetics , RNA Processing, Post-Transcriptional/drug effects , RNA Splicing , RNA, Small Nuclear/genetics , Simian virus 40/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...