Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Drug Discov Today ; 28(1): 103431, 2023 01.
Article in English | MEDLINE | ID: mdl-36356786

ABSTRACT

The concept of using small molecules to therapeutically modulate pre-mRNA splicing was validated with the US Food and Drug Administration (FDA) approval of Evrysdi® (risdiplam) in 2020. Since then, efforts have continued unabated toward the discovery of new splicing-modulating drugs. However, the drug development world has evolved in the 10 years since risdiplam precursors were first identified in high-throughput screening (HTS). Now, new mechanistic insights into RNA-processing pathways and regulatory networks afford increasingly feasible targeted approaches. In this review, organized into classes of biological target, we compile and summarize small molecules discovered, devised, and developed since 2020 to alter pre-mRNA splicing.


Subject(s)
RNA Precursors , RNA Splicing , RNA Precursors/genetics , RNA Precursors/metabolism , Azo Compounds , Pyrimidines , Alternative Splicing
2.
Front Oncol ; 12: 832816, 2022.
Article in English | MEDLINE | ID: mdl-35223511

ABSTRACT

Blocking the pyrimidine nucleotide de novo synthesis pathway by inhibiting dihydroorotate dehydrogenase (DHODH) results in the cell cycle arrest and/or differentiation of rapidly proliferating cells including activated lymphocytes, cancer cells, or virally infected cells. Emvododstat (PTC299) is an orally bioavailable small molecule that inhibits DHODH. We evaluated the potential for emvododstat to inhibit the progression of acute myeloid leukemia (AML) using several in vitro and in vivo models of the disease. Broad potent activity was demonstrated against multiple AML cell lines, AML blasts cultured ex vivo from patient blood samples, and AML tumor models including patient-derived xenograft models. Emvododstat induced differentiation, cytotoxicity, or both in primary AML patient blasts cultured ex vivo with 8 of 10 samples showing sensitivity. AML cells with diverse driver mutations were sensitive, suggesting the potential of emvododstat for broad therapeutic application. AML cell lines that are not sensitive to emvododstat are likely to be more reliant on the salvage pathway than on de novo synthesis of pyrimidine nucleotides. Pharmacokinetic experiments in rhesus monkeys demonstrated that emvododstat levels rose rapidly after oral administration, peaking about 2 hours post-dosing. This was associated with an increase in the levels of dihydroorotate (DHO), the substrate for DHODH, within 2 hours of dosing indicating that DHODH inhibition is rapid. DHO levels declined as drug levels declined, consistent with the reversibility of DHODH inhibition by emvododstat. These preclinical findings provide a rationale for clinical evaluation of emvododstat in an ongoing Phase 1 study of patients with relapsed/refractory acute leukemias.

3.
Nat Commun ; 12(1): 7299, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34911927

ABSTRACT

Huntington's disease (HD) is a hereditary neurodegenerative disorder caused by expansion of cytosine-adenine-guanine (CAG) trinucleotide repeats in the huntingtin (HTT) gene. Consequently, the mutant protein is ubiquitously expressed and drives pathogenesis of HD through a toxic gain-of-function mechanism. Animal models of HD have demonstrated that reducing huntingtin (HTT) protein levels alleviates motor and neuropathological abnormalities. Investigational drugs aim to reduce HTT levels by repressing HTT transcription, stability or translation. These drugs require invasive procedures to reach the central nervous system (CNS) and do not achieve broad CNS distribution. Here, we describe the identification of orally bioavailable small molecules with broad distribution throughout the CNS, which lower HTT expression consistently throughout the CNS and periphery through selective modulation of pre-messenger RNA splicing. These compounds act by promoting the inclusion of a pseudoexon containing a premature termination codon (stop-codon psiExon), leading to HTT mRNA degradation and reduction of HTT levels.


Subject(s)
Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Huntington Disease/drug therapy , Huntington Disease/genetics , RNA Splicing , Small Molecule Libraries/administration & dosage , Animals , Central Nervous System/drug effects , Central Nervous System/metabolism , Disease Models, Animal , Humans , Huntington Disease/metabolism , Mice , RNA Splicing/drug effects , RNA Stability/drug effects , Trinucleotide Repeat Expansion/drug effects
4.
Nat Commun ; 12(1): 3332, 2021 06 07.
Article in English | MEDLINE | ID: mdl-34099697

ABSTRACT

Pre-mRNA splicing is a key controller of human gene expression. Disturbances in splicing due to mutation lead to dysregulated protein expression and contribute to a substantial fraction of human disease. Several classes of splicing modulator compounds (SMCs) have been recently identified and establish that pre-mRNA splicing represents a target for therapy. We describe herein the identification of BPN-15477, a SMC that restores correct splicing of ELP1 exon 20. Using transcriptome sequencing from treated fibroblast cells and a machine learning approach, we identify BPN-15477 responsive sequence signatures. We then leverage this model to discover 155 human disease genes harboring ClinVar mutations predicted to alter pre-mRNA splicing as targets for BPN-15477. Splicing assays confirm successful correction of splicing defects caused by mutations in CFTR, LIPA, MLH1 and MAPT. Subsequent validations in two disease-relevant cellular models demonstrate that BPN-15477 increases functional protein, confirming the clinical potential of our predictions.


Subject(s)
Deep Learning , Gene Targeting/methods , RNA Splicing , Animals , Computational Biology , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Exons , HEK293 Cells , Humans , Mice , Mice, Transgenic , MutL Protein Homolog 1/genetics , Mutation , Phenethylamines/administration & dosage , Pyridazines/administration & dosage , Sterol Esterase/genetics , Transcriptome , tau Proteins/genetics
5.
Virus Res ; 292: 198246, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33249060

ABSTRACT

The coronavirus disease 2019 (COVID-19) pandemic has created an urgent need for therapeutics that inhibit the SARS-COV-2 virus and suppress the fulminant inflammation characteristic of advanced illness. Here, we describe the anti-COVID-19 potential of PTC299, an orally bioavailable compound that is a potent inhibitor of dihydroorotate dehydrogenase (DHODH), the rate-limiting enzyme of the de novo pyrimidine nucleotide biosynthesis pathway. In tissue culture, PTC299 manifests robust, dose-dependent, and DHODH-dependent inhibition of SARS-COV-2 replication (EC50 range, 2.0-31.6 nM) with a selectivity index >3,800. PTC299 also blocked replication of other RNA viruses, including Ebola virus. Consistent with known DHODH requirements for immunomodulatory cytokine production, PTC299 inhibited the production of interleukin (IL)-6, IL-17A (also called IL-17), IL-17 F, and vascular endothelial growth factor (VEGF) in tissue culture models. The combination of anti-SARS-CoV-2 activity, cytokine inhibitory activity, and previously established favorable pharmacokinetic and human safety profiles render PTC299 a promising therapeutic for COVID-19.


Subject(s)
Antiviral Agents/pharmacology , Carbamates/pharmacology , Carbazoles/pharmacology , Cytokines/antagonists & inhibitors , Oxidoreductases Acting on CH-CH Group Donors/antagonists & inhibitors , SARS-CoV-2/drug effects , Virus Replication/drug effects , Animals , Chlorocebus aethiops , Cytokine Release Syndrome/drug therapy , Cytokines/immunology , Dihydroorotate Dehydrogenase , HeLa Cells , Humans , Inflammation/drug therapy , Inflammation/virology , Vero Cells , COVID-19 Drug Treatment
6.
bioRxiv ; 2020 Aug 05.
Article in English | MEDLINE | ID: mdl-32793904

ABSTRACT

The coronavirus disease 2019 (COVID-19) pandemic has created an urgent need for therapeutics that inhibit the SARS-CoV-2 virus and suppress the fulminant inflammation characteristic of advanced illness. Here, we describe the anti-COVID-19 potential of PTC299, an orally available compound that is a potent inhibitor of dihydroorotate dehydrogenase (DHODH), the rate-limiting enzyme of the de novo pyrimidine biosynthesis pathway. In tissue culture, PTC299 manifests robust, dose-dependent, and DHODH-dependent inhibition of SARS CoV-2 replication (EC 50 range, 2.0 to 31.6 nM) with a selectivity index >3,800. PTC299 also blocked replication of other RNA viruses, including Ebola virus. Consistent with known DHODH requirements for immunomodulatory cytokine production, PTC299 inhibited the production of interleukin (IL)-6, IL-17A (also called IL-17), IL-17F, and vascular endothelial growth factor (VEGF) in tissue culture models. The combination of anti-SARS-CoV-2 activity, cytokine inhibitory activity, and previously established favorable pharmacokinetic and human safety profiles render PTC299 a promising therapeutic for COVID-19.

7.
Mol Cancer Ther ; 18(1): 3-16, 2019 01.
Article in English | MEDLINE | ID: mdl-30352802

ABSTRACT

PTC299 was identified as an inhibitor of VEGFA mRNA translation in a phenotypic screen and evaluated in the clinic for treatment of solid tumors. To guide precision cancer treatment, we performed extensive biological characterization of the activity of PTC299 and demonstrated that inhibition of VEGF production and cell proliferation by PTC299 is linked to a decrease in uridine nucleotides by targeting dihydroorotate dehydrogenase (DHODH), a rate-limiting enzyme for de novo pyrimidine nucleotide synthesis. Unlike previously reported DHODH inhibitors that were identified using in vitro enzyme assays, PTC299 is a more potent inhibitor of DHODH in isolated mitochondria suggesting that mitochondrial membrane lipid engagement in the DHODH conformation in situ is required for its optimal activity. PTC299 has broad and potent activity against hematologic cancer cells in preclinical models, reflecting a reduced pyrimidine nucleotide salvage pathway in leukemia cells. Archived serum samples from patients treated with PTC299 demonstrated increased levels of dihydroorotate, the substrate of DHODH, indicating target engagement in patients. PTC299 has advantages over previously reported DHODH inhibitors, including greater potency, good oral bioavailability, and lack of off-target kinase inhibition and myelosuppression, and thus may be useful for the targeted treatment of hematologic malignancies.


Subject(s)
Hematologic Neoplasms/drug therapy , Imidazoles/administration & dosage , Oxidoreductases Acting on CH-CH Group Donors/antagonists & inhibitors , Thiazoles/administration & dosage , Vascular Endothelial Growth Factor A/genetics , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dihydroorotate Dehydrogenase , Hematologic Neoplasms/blood , Hematologic Neoplasms/enzymology , Humans , Imidazoles/pharmacology , K562 Cells , Mice , Oxidoreductases Acting on CH-CH Group Donors/blood , Thiazoles/pharmacology , Xenograft Model Antitumor Assays
8.
RNA ; 23(4): 567-577, 2017 04.
Article in English | MEDLINE | ID: mdl-28096517

ABSTRACT

Nonsense mutations resulting in a premature stop codon in an open reading frame occur in critical tumor suppressor genes in a large number of the most common forms of cancers and are known to cause or contribute to the progression of disease. Low molecular weight compounds that induce readthrough of nonsense mutations offer a new means of treating patients with genetic disorders or cancers resulting from nonsense mutations. We have identified the nucleoside analog clitocine as a potent and efficacious suppressor of nonsense mutations. We determined that incorporation of clitocine into RNA during transcription is a prerequisite for its readthrough activity; the presence of clitocine in the third position of a premature stop codon directly induces readthrough. We demonstrate that clitocine can induce the production of p53 protein in cells harboring p53 nonsense-mutated alleles. In these cells, clitocine restored production of full-length and functional p53 as evidenced by induced transcriptional activation of downstream p53 target genes, progression of cells into apoptosis, and impeded growth of nonsense-containing human ovarian cancer tumors in xenograft tumor models. Thus, clitocine induces readthrough of nonsense mutations by a previously undescribed mechanism and represents a novel therapeutic modality to treat cancers and genetic diseases caused by nonsense mutations.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Biomimetic Materials/pharmacology , Codon, Nonsense/drug effects , Furans/pharmacology , Nucleosides/pharmacology , Ovarian Neoplasms/drug therapy , Pyrimidine Nucleosides/pharmacology , Tumor Suppressor Protein p53/agonists , Animals , Antimetabolites, Antineoplastic/chemical synthesis , Antimetabolites, Antineoplastic/metabolism , Apoptosis/drug effects , Biomimetic Materials/chemical synthesis , Biomimetic Materials/metabolism , Cell Line, Tumor , Female , Furans/chemical synthesis , Furans/metabolism , Genes, Reporter , Humans , Luciferases/genetics , Luciferases/metabolism , Mice , Mice, Nude , Nucleosides/chemical synthesis , Nucleosides/metabolism , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Protein Biosynthesis , Pyrimidine Nucleosides/chemical synthesis , Pyrimidine Nucleosides/metabolism , Signal Transduction , Transcriptional Activation , Tumor Burden/drug effects , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays
9.
PLoS One ; 11(12): e0168366, 2016.
Article in English | MEDLINE | ID: mdl-27992500

ABSTRACT

Current anti-VEGF (Vascular Endothelial Growth Factor A) therapies to treat various cancers indiscriminately block VEGF function in the patient resulting in the global loss of VEGF signaling which has been linked to dose-limiting toxicities as well as treatment failures due to acquired resistance. Accumulating evidence suggests that this resistance is at least partially due to increased production of compensatory tumor angiogenic factors/cytokines. VEGF protein production is differentially controlled depending on whether cells are in the normal "homeostatic" state or in a stressed state, such as hypoxia, by post-transcriptional regulation imparted by elements in the 5' and 3' untranslated regions (UTR) of the VEGF mRNA. Using the Gene Expression Modulation by Small molecules (GEMS™) phenotypic assay system, we performed a high throughput screen to identify low molecular weight compounds that target the VEGF mRNA UTR-mediated regulation of stress-induced VEGF production in tumor cells. We identified a number of compounds that potently and selectively reduce endogenous VEGF production under hypoxia in HeLa cells. Medicinal chemistry efforts improved the potency and pharmaceutical properties of one series of compounds resulting in the discovery of PTC-510 which inhibits hypoxia-induced VEGF expression in HeLa cells at low nanomolar concentration. In mouse xenograft studies, oral administration of PTC-510 results in marked reduction of intratumor VEGF production and single agent control of tumor growth without any evident toxicity. Here, we show that selective suppression of stress-induced VEGF production within tumor cells effectively controls tumor growth. Therefore, this approach may minimize the liabilities of current global anti-VEGF therapies.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Antineoplastic Agents/administration & dosage , High-Throughput Screening Assays/methods , Neoplasms/drug therapy , Untranslated Regions/drug effects , Vascular Endothelial Growth Factor A/genetics , Administration, Oral , Angiogenesis Inhibitors/pharmacology , Animals , Antineoplastic Agents/pharmacology , Cell Hypoxia , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , HEK293 Cells , HeLa Cells , Hep G2 Cells , Humans , Mice , Neoplasms/genetics , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Xenograft Model Antitumor Assays
10.
Proc Natl Acad Sci U S A ; 113(44): 12508-12513, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27702906

ABSTRACT

A premature termination codon (PTC) in the ORF of an mRNA generally leads to production of a truncated polypeptide, accelerated degradation of the mRNA, and depression of overall mRNA expression. Accordingly, nonsense mutations cause some of the most severe forms of inherited disorders. The small-molecule drug ataluren promotes therapeutic nonsense suppression and has been thought to mediate the insertion of near-cognate tRNAs at PTCs. However, direct evidence for this activity has been lacking. Here, we expressed multiple nonsense mutation reporters in human cells and yeast and identified the amino acids inserted when a PTC occupies the ribosomal A site in control, ataluren-treated, and aminoglycoside-treated cells. We find that ataluren's likely target is the ribosome and that it produces full-length protein by promoting insertion of near-cognate tRNAs at the site of the nonsense codon without apparent effects on transcription, mRNA processing, mRNA stability, or protein stability. The resulting readthrough proteins retain function and contain amino acid replacements similar to those derived from endogenous readthrough, namely Gln, Lys, or Tyr at UAA or UAG PTCs and Trp, Arg, or Cys at UGA PTCs. These insertion biases arise primarily from mRNA:tRNA mispairing at codon positions 1 and 3 and reflect, in part, the preferred use of certain nonstandard base pairs, e.g., U-G. Ataluren's retention of similar specificity of near-cognate tRNA insertion as occurs endogenously has important implications for its general use in therapeutic nonsense suppression.


Subject(s)
Codon, Nonsense/genetics , Oxadiazoles/pharmacology , RNA, Transfer/genetics , Ribosomes/drug effects , HEK293 Cells , Humans , Protein Biosynthesis/drug effects , RNA Stability/drug effects , RNA, Transfer/metabolism , Ribosomes/genetics , Ribosomes/metabolism , Transcription, Genetic/drug effects
12.
Am J Hum Genet ; 99(1): 228-35, 2016 Jul 07.
Article in English | MEDLINE | ID: mdl-27392077

ABSTRACT

The tRNA splicing endonuclease is a highly evolutionarily conserved protein complex, involved in the cleavage of intron-containing tRNAs. In human it consists of the catalytic subunits TSEN2 and TSEN34, as well as the non-catalytic TSEN54 and TSEN15. Recessive mutations in the corresponding genes of the first three are known to cause pontocerebellar hypoplasia (PCH) types 2A-C, 4, and 5. Here, we report three homozygous TSEN15 variants that cause a milder version of PCH2. The affected individuals showed progressive microcephaly, delayed developmental milestones, intellectual disability, and, in two out of four cases, epilepsy. None, however, displayed the central visual failure seen in PCH case subjects where other subunits of the TSEN are mutated, and only one was affected by the extensive motor defects that are typical in other forms of PCH2. The three amino acid substitutions impacted the protein level of TSEN15 and the stoichiometry of the interacting subunits in different ways, but all resulted in an almost complete loss of in vitro tRNA cleavage activity. Taken together, our results demonstrate that mutations in any known subunit of the TSEN complex can cause PCH and progressive microcephaly, emphasizing the importance of its function during brain development.


Subject(s)
Cerebellar Diseases/genetics , Endonucleases/genetics , Genes, Recessive , Microcephaly/genetics , Mutation , Amino Acid Sequence , Child , Child, Preschool , Endonucleases/chemistry , Female , Humans , Infant , Infant, Newborn , Male , Models, Molecular , Pedigree
13.
J Biol Chem ; 290(26): 16021-30, 2015 Jun 26.
Article in English | MEDLINE | ID: mdl-25971974

ABSTRACT

The tRNA splicing endonuclease (Sen) complex is located on the mitochondrial outer membrane and splices precursor tRNAs in Saccharomyces cerevisiae. Here, we demonstrate that the Sen complex cleaves the mitochondria-localized mRNA encoding Cbp1 (cytochrome b mRNA processing 1). Endonucleolytic cleavage of this mRNA required two cis-elements: the mitochondrial targeting signal and the stem-loop 652-726-nt region. Mitochondrial localization of the Sen complex was required for cleavage of the CBP1 mRNA, and the Sen complex cleaved this mRNA directly in vitro. We propose that the Sen complex cleaves the CBP1 mRNA, which is co-translationally localized to mitochondria via its mitochondrial targeting signal.


Subject(s)
Endoribonucleases/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/genetics , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/enzymology , Base Sequence , Endoribonucleases/genetics , Mitochondria/genetics , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/metabolism , Molecular Sequence Data , Nucleic Acid Conformation , RNA Splicing , RNA, Messenger/chemistry , RNA, Messenger/metabolism , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism
14.
Cell ; 157(3): 651-63, 2014 Apr 24.
Article in English | MEDLINE | ID: mdl-24766810

ABSTRACT

Neurodegenerative diseases can occur so early as to affect neurodevelopment. From a cohort of more than 2,000 consanguineous families with childhood neurological disease, we identified a founder mutation in four independent pedigrees in cleavage and polyadenylation factor I subunit 1 (CLP1). CLP1 is a multifunctional kinase implicated in tRNA, mRNA, and siRNA maturation. Kinase activity of the CLP1 mutant protein was defective, and the tRNA endonuclease complex (TSEN) was destabilized, resulting in impaired pre-tRNA cleavage. Germline clp1 null zebrafish showed cerebellar neurodegeneration that was rescued by wild-type, but not mutant, human CLP1 expression. Patient-derived induced neurons displayed both depletion of mature tRNAs and accumulation of unspliced pre-tRNAs. Transfection of partially processed tRNA fragments into patient cells exacerbated an oxidative stress-induced reduction in cell survival. Our data link tRNA maturation to neuronal development and neurodegeneration through defective CLP1 function in humans.


Subject(s)
Cerebellum/growth & development , Cerebellum/pathology , Cleavage And Polyadenylation Specificity Factor/metabolism , Nuclear Proteins/genetics , Phosphotransferases/genetics , RNA Splicing , RNA, Transfer/genetics , Transcription Factors/genetics , Zebrafish Proteins/metabolism , Animals , Brain/metabolism , Brain/pathology , Cleavage And Polyadenylation Specificity Factor/genetics , Female , Humans , Male , Mice , Models, Molecular , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Nuclear Proteins/metabolism , Pedigree , Phosphotransferases/metabolism , RNA, Transfer/metabolism , Saccharomyces cerevisiae/metabolism , Transcription Factors/metabolism , Zebrafish , Zebrafish Proteins/genetics
15.
Mol Cell ; 52(2): 184-92, 2013 Oct 24.
Article in English | MEDLINE | ID: mdl-24095278

ABSTRACT

In cells, tRNAs are synthesized as precursor molecules bearing extra sequences at their 5' and 3' ends. Some tRNAs also contain introns, which, in archaea and eukaryotes, are cleaved by an evolutionarily conserved endonuclease complex that generates fully functional mature tRNAs. In addition, tRNAs undergo numerous posttranscriptional nucleotide chemical modifications. In Trypanosoma brucei, the single intron-containing tRNA (tRNA(Tyr)GUA) is responsible for decoding all tyrosine codons; therefore, intron removal is essential for viability. Using molecular and biochemical approaches, we show the presence of several noncanonical editing events, within the intron of pre-tRNA(Tyr)GUA, involving guanosine-to-adenosine transitions (G to A) and an adenosine-to-uridine transversion (A to U). The RNA editing described here is required for proper processing of the intron, establishing the functional significance of noncanonical editing with implications for tRNA processing in the deeply divergent kinetoplastid lineage and eukaryotes in general.


Subject(s)
Introns/genetics , RNA Editing , RNA Splicing , RNA, Transfer, Tyr/genetics , Trypanosoma brucei brucei/genetics , Amino Acid Sequence , Base Sequence , Blotting, Northern , Endoribonucleases/genetics , Endoribonucleases/metabolism , Molecular Sequence Data , Nucleic Acid Conformation , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , RNA Interference , RNA Precursors/genetics , RNA Precursors/metabolism , RNA Processing, Post-Transcriptional , RNA, Protozoan/genetics , RNA, Protozoan/metabolism , RNA, Transfer, Tyr/chemistry , RNA, Transfer, Tyr/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Trypanosoma brucei brucei/metabolism
16.
RNA Biol ; 6(3): 329-34, 2009.
Article in English | MEDLINE | ID: mdl-19574739

ABSTRACT

Post-transcriptional regulatory mechanisms, dependent on specific RNA:RNA, RNA:protein, or protein:protein interactions that generate large numbers of different RNP constellations, can have sizeable effects on the expression of any given gene. At the mRNA-specific level, these mechanisms also provide numerous novel targets for small molecule drugs capable of enhancing or inhibiting the accumulation of specific proteins. Here, we describe two drug screening technologies that target the post-transcriptional regulation of specific mRNAs with specific small molecules. In one case the GEMS technology utilizes mRNA-specific 5'- and 3'-UTR pairs to identify compounds that reduce protein production as a consequence of the UTRs. The second example utilizes nonsense-containing mRNAs to identify compounds capable of promoting therapeutic nonsense suppression. Both programs have yielded drug candidates that are presently in clinical testing for human diseases with high unmet clinical needs, thus illustrating the therapeutic potential of targeting post-transcriptional control.


Subject(s)
Drug Evaluation, Preclinical , Gene Expression Regulation , RNA, Messenger/genetics , Animals , Humans , RNA Stability , RNA, Messenger/metabolism , Transcription, Genetic
18.
Drug Discov Today ; 12(13-14): 553-60, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17631250

ABSTRACT

The physiological levels of many clinically important proteins are regulated through cellular mechanisms that control the stability and translational efficiency of mRNA. These post-transcriptional processes, which play a critical role in the regulation of gene expression, depend on interactions of specific trans-acting factors with sequence elements located within the 5'- and 3'-untranslated regions (UTRs) of an mRNA. A technology platform called GEMS (Gene Expression Modulation by Small-molecules) exploits the interactions of UTR elements with the trans-acting factors, thereby specifically targeting mechanisms of post-transcriptional control. In this review we describe how this technology enables the identification of small-molecules that modulate the levels of proteins involved in disease pathogenesis.


Subject(s)
Drug Design , Gene Expression Regulation , RNA Processing, Post-Transcriptional , Small Molecule Libraries , 3' Untranslated Regions , 5' Untranslated Regions , Humans , RNA, Messenger/genetics , RNA, Messenger/physiology , Trans-Activators/genetics , Trans-Activators/metabolism
20.
Nature ; 447(7140): 87-91, 2007 May 03.
Article in English | MEDLINE | ID: mdl-17450125

ABSTRACT

Nonsense mutations promote premature translational termination and cause anywhere from 5-70% of the individual cases of most inherited diseases. Studies on nonsense-mediated cystic fibrosis have indicated that boosting specific protein synthesis from <1% to as little as 5% of normal levels may greatly reduce the severity or eliminate the principal manifestations of disease. To address the need for a drug capable of suppressing premature termination, we identified PTC124-a new chemical entity that selectively induces ribosomal readthrough of premature but not normal termination codons. PTC124 activity, optimized using nonsense-containing reporters, promoted dystrophin production in primary muscle cells from humans and mdx mice expressing dystrophin nonsense alleles, and rescued striated muscle function in mdx mice within 2-8 weeks of drug exposure. PTC124 was well tolerated in animals at plasma exposures substantially in excess of those required for nonsense suppression. The selectivity of PTC124 for premature termination codons, its well characterized activity profile, oral bioavailability and pharmacological properties indicate that this drug may have broad clinical potential for the treatment of a large group of genetic disorders with limited or no therapeutic options.


Subject(s)
Codon, Nonsense/genetics , Genetic Diseases, Inborn/drug therapy , Genetic Diseases, Inborn/genetics , Oxadiazoles/pharmacology , Oxadiazoles/therapeutic use , Protein Biosynthesis/drug effects , Alleles , Animals , Biological Availability , Dystrophin/biosynthesis , Dystrophin/genetics , Genetic Diseases, Inborn/blood , Humans , Mice , Mice, Inbred mdx , Oxadiazoles/administration & dosage , Oxadiazoles/pharmacokinetics , Phenotype , Protein Biosynthesis/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...