Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
Add more filters










Publication year range
1.
Cell Chem Biol ; 28(8): 1145-1157.e6, 2021 08 19.
Article in English | MEDLINE | ID: mdl-33689684

ABSTRACT

Dysregulated pre-mRNA splicing is an emerging Achilles heel of cancers and myelodysplasias. To expand the currently limited portfolio of small-molecule drug leads, we screened for chemical modulators of the U2AF complex, which nucleates spliceosome assembly and is mutated in myelodysplasias. A hit compound specifically enhances RNA binding by a U2AF2 subunit. Remarkably, the compound inhibits splicing of representative substrates and stalls spliceosome assembly at the stage of U2AF function. Computational docking, together with structure-guided mutagenesis, indicates that the compound bridges the tandem U2AF2 RNA recognition motifs via hydrophobic and electrostatic moieties. Cells expressing a cancer-associated U2AF1 mutant are preferentially killed by treatment with the compound. Altogether, our results highlight the potential of trapping early spliceosome assembly as an effective pharmacological means to manipulate pre-mRNA splicing. By extension, we suggest that stabilizing assembly intermediates may offer a useful approach for small-molecule inhibition of macromolecular machines.


Subject(s)
RNA Precursors/drug effects , RNA Splicing/drug effects , RNA, Neoplasm/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Splicing Factor U2AF/antagonists & inhibitors , Female , HEK293 Cells , Humans , K562 Cells , Molecular Docking Simulation , Molecular Structure , RNA Precursors/genetics , RNA Splicing/genetics , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Splicing Factor U2AF/genetics , Splicing Factor U2AF/metabolism
2.
ACS Chem Biol ; 15(4): 808-818, 2020 04 17.
Article in English | MEDLINE | ID: mdl-32191432

ABSTRACT

RNA splicing is a key component of gene expression and proteomic diversity in humans. The spliceosome assembles on and processes individual nascent pre-mRNA transcripts into distinct mature mRNAs that can code for different proteins. Splicing programs can be affected by somatic mutations and changes in response to exogenous stimuli. Importantly, alterations in splicing can be direct drivers of diseases including cancers. This Review describes recent advances and the potential for targeting and controlling pre-mRNA splicing in humans with small molecules, ranging from targeting spliceosomal proteins to direct targeting of individual RNA transcripts.


Subject(s)
Protein Kinase Inhibitors/pharmacology , RNA Precursors/metabolism , RNA Splicing/drug effects , G-Quadruplexes/drug effects , Humans , RNA Precursors/drug effects , Serine-Arginine Splicing Factors/metabolism , Spliceosomes/drug effects
3.
Nat Rev Drug Discov ; 19(2): 112-129, 2020 02.
Article in English | MEDLINE | ID: mdl-31554928

ABSTRACT

Discoveries in the past decade have highlighted the potential of mRNA as a therapeutic target for cancer. Specifically, RNA sequencing revealed that, in addition to gene mutations, alterations in mRNA can contribute to the initiation and progression of cancer. Indeed, precursor mRNA processing, which includes the removal of introns by splicing and the formation of 3' ends by cleavage and polyadenylation, is frequently altered in tumours. These alterations result in numerous cancer-specific mRNAs that generate altered levels of normal proteins or proteins with new functions, leading to the activation of oncogenes or the inactivation of tumour-suppressor genes. Abnormally spliced and polyadenylated mRNAs are also associated with resistance to cancer treatment and, unexpectedly, certain cancers are highly sensitive to the pharmacological inhibition of splicing. This Review summarizes recent progress in our understanding of how splicing and polyadenylation are altered in cancer and highlights how this knowledge has been translated for drug discovery, resulting in the production of small molecules and oligonucleotides that modulate the spliceosome and are in clinical trials for the treatment of cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , RNA Precursors/drug effects , RNA Processing, Post-Transcriptional/drug effects , RNA Splicing/drug effects , RNA, Messenger/genetics , Humans , Molecular Targeted Therapy , Neoplasms/genetics
4.
Int J Mol Sci ; 20(20)2019 Oct 11.
Article in English | MEDLINE | ID: mdl-31614438

ABSTRACT

The process of pre-mRNA splicing is a common and fundamental step in the expression of most human genes. Alternative splicing, whereby different splice motifs and sites are recognised in a developmental and/or tissue-specific manner, contributes to genetic plasticity and diversity of gene expression. Redirecting pre-mRNA processing of various genes has now been validated as a viable clinical therapeutic strategy, providing treatments for Duchenne muscular dystrophy (inducing specific exon skipping) and spinal muscular atrophy (promoting exon retention). We have designed and evaluated over 5000 different antisense oligonucleotides to alter splicing of a variety of pre-mRNAs, from the longest known human pre-mRNA to shorter, exon-dense primary gene transcripts. Here, we present our guidelines for designing, evaluating and optimising splice switching antisense oligomers in vitro. These systematic approaches assess several critical factors such as the selection of target splicing motifs, choice of cells, various delivery reagents and crucial aspects of validating assays for the screening of antisense oligonucleotides composed of 2'-O-methyl modified bases on a phosphorothioate backbone.


Subject(s)
Alternative Splicing/drug effects , Oligonucleotides, Antisense/chemical synthesis , RNA Precursors/genetics , Animals , Cell Line , Drug Design , Guidelines as Topic , HEK293 Cells , Humans , Oligonucleotides, Antisense/chemistry , Oligonucleotides, Antisense/pharmacology , RNA Precursors/drug effects
6.
Cancer Cell ; 33(3): 386-400.e5, 2018 03 12.
Article in English | MEDLINE | ID: mdl-29478914

ABSTRACT

To identify novel targets for acute myeloid leukemia (AML) therapy, we performed genome-wide CRISPR-Cas9 screening using AML cell lines, followed by a second screen in vivo. Here, we show that the mRNA decapping enzyme scavenger (DCPS) gene is essential for AML cell survival. The DCPS enzyme interacted with components of pre-mRNA metabolic pathways, including spliceosomes, as revealed by mass spectrometry. RG3039, a DCPS inhibitor originally developed to treat spinal muscular atrophy, exhibited anti-leukemic activity via inducing pre-mRNA mis-splicing. Humans harboring germline biallelic DCPS loss-of-function mutations do not exhibit aberrant hematologic phenotypes, indicating that DCPS is dispensable for human hematopoiesis. Our findings shed light on a pre-mRNA metabolic pathway and identify DCPS as a target for AML therapy.


Subject(s)
CRISPR-Cas Systems/drug effects , Endoribonucleases/drug effects , Leukemia/drug therapy , Muscular Atrophy, Spinal/drug therapy , Quinazolines/pharmacology , Animals , CRISPR-Cas Systems/genetics , Cell Line , Endoribonucleases/genetics , Endoribonucleases/metabolism , Humans , Leukemia/genetics , Male , Metabolic Networks and Pathways/drug effects , Mice, Inbred C57BL , Muscular Atrophy, Spinal/genetics , RNA Precursors/drug effects , RNA Precursors/genetics , RNA Splicing/drug effects , RNA Splicing/genetics , RNA, Messenger/genetics
7.
J Med Chem ; 59(22): 10190-10197, 2016 11 23.
Article in English | MEDLINE | ID: mdl-27753493

ABSTRACT

U2AF homology motifs (UHMs) are atypical RNA recognition motif domains that mediate critical protein-protein interactions during the regulation of alternative pre-mRNA splicing and other processes. The recognition of UHM domains by UHM ligand motif (ULM) peptide sequences plays important roles during early steps of spliceosome assembly. Splicing factor 45 kDa (SPF45) is an alternative splicing factor implicated in breast and lung cancers, and splicing regulation of apoptosis-linked pre-mRNAs by SPF45 was shown to depend on interactions between its UHM domain and ULM motifs in constitutive splicing factors. We have developed cyclic peptide inhibitors that target UHM domains. By screening a focused library of linear and cyclic peptides and performing structure-activity relationship analysis, we designed cyclic peptides with 4-fold improved binding affinity for the SPF45 UHM domain compared to native ULM ligands and 270-fold selectivity to discriminate UHM domains from alternative and constitutive splicing factors. These inhibitors are useful tools to modulate and dissect mechanisms of alternative splicing regulation.


Subject(s)
Drug Design , Peptides, Cyclic/pharmacology , RNA Precursors/drug effects , RNA Splicing/drug effects , Splicing Factor U2AF/antagonists & inhibitors , Dose-Response Relationship, Drug , Humans , Molecular Structure , Peptides, Cyclic/chemical synthesis , Peptides, Cyclic/chemistry , Splicing Factor U2AF/metabolism , Structure-Activity Relationship
8.
J Nat Prod ; 76(4): 685-93, 2013 Apr 26.
Article in English | MEDLINE | ID: mdl-23517093

ABSTRACT

Mining the genome sequence of Burkholderia thailandensis MSMB43 revealed a cryptic biosynthetic gene cluster resembling that of FR901464 (4), a prototype spliceosome inhibitor produced by Pseudomonas sp. No. 2663. Transcriptional analysis revealed a cultivation condition in which a regulatory gene of the cryptic gene cluster is adequately expressed. Consequently, three new compounds, named thailanstatins A (1), B (2), and C (3), were isolated from the fermentation broth of B. thailandensis MSMB43. Thailanstatins are proposed to be biosynthesized by a hybrid polyketide synthase-nonribosomal peptide synthetase pathway. They differ from 4 by lacking an unstable hydroxyl group and by having an extra carboxyl moiety; those differences endow thailanstatins with a significantly greater stability than 4 as tested in phosphate buffer at pH 7.4. In vitro assays showed that thailanstatins inhibit pre-mRNA splicing as potently as 4, with half-maximal inhibitory concentrations in the single to sub-µM range. Cell culture assays indicated that thailanstatins also possess potent antiproliferative activities in representative human cancer cell lines, with half-maximal growth inhibitory concentrations in the single nM range. This work provides new chemical entities for research and development and new structure-activity information for chemical optimization of related spliceosome inhibitors.


Subject(s)
Antineoplastic Agents/isolation & purification , Antineoplastic Agents/pharmacology , Burkholderia/chemistry , Pyrans/isolation & purification , Pyrans/pharmacology , Antineoplastic Agents/chemistry , Drug Screening Assays, Antitumor , Genomics , Humans , Multigene Family , Pseudomonas/chemistry , Pyrans/chemistry , RNA Precursors/drug effects , Spiro Compounds/chemistry , Spiro Compounds/isolation & purification , Structure-Activity Relationship
9.
Chemistry ; 17(3): 895-904, 2011 Jan 17.
Article in English | MEDLINE | ID: mdl-21226105

ABSTRACT

FR901464, a natural product isolated from a bacterium source, activates a reporter gene, inhibits pre-mRNA splicing, and shows antitumor activity. We previously reported the development of a more potent analogue, meayamycin, through the total synthesis of FR901464. Herein, we report detailed structure-activity relationships of FR901464 that revealed the significance of the epoxide, carbon atoms in the tetrahydropyran ring, the Z geometry of the side chain, the 1,3-diene moiety, the C4-hydroxy group, and the C2''-carbonyl group. Importantly, the methyl group of the acetyl substituent was found to be inessential, leading to a new potent analogue. Additionally, partially based on in vivo data, we synthesized and evaluated potentially more metabolically stable analogues for their antiproliferative activity. These structural insights into FR901464 may contribute to the simplification of the natural product for further drug development.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Biological Products/chemistry , Biological Products/pharmacology , Cell Line, Tumor/drug effects , Cell Proliferation/drug effects , Epoxy Compounds/chemistry , Epoxy Compounds/pharmacology , Pyrans/chemistry , Pyrans/pharmacology , RNA Precursors/drug effects , RNA Splicing/drug effects , Animals , Humans , Mice , Molecular Structure , Spiro Compounds/chemistry , Spiro Compounds/pharmacology , Stereoisomerism , Structure-Activity Relationship
10.
ACS Chem Biol ; 6(1): 21-33, 2011 Jan 21.
Article in English | MEDLINE | ID: mdl-21105689

ABSTRACT

The identification of the mechanism of action and therapeutic potential of bioactive small molecules remains a considerable challenge in the field of drug discovery and chemical biology. Apart from traditional target identification techniques, new tools have emerged that can significantly aid mechanism elucidation efforts. The development of pattern matching algorithms that compare transcription profile data to analogous data on compounds with known cellular targets allows for mechanistic insights without the need to synthesize chemically modified probes. In addition, such methods can be used to connect small molecules to particular disease states, thus aiding the rational identification of candidate therapeutics. Another method with considerable potential is whole-genome RNAi screening, a technique that can identify critical upstream proteins involved in a small molecule's mechanism of action. Several proof-of-concept studies using compounds with known cellular targets suggest this tool will enable mechanistic characterization of bioactive small molecules with unknown mechanisms. This Review highlights recent successes in using these pattern matching and chemical genetic tools, with the goal of uncovering small molecule mechanisms and identifying therapeutic candidates for disease treatment.


Subject(s)
Drug Design , Protein Array Analysis/methods , RNA Interference/drug effects , RNA Precursors/drug effects , RNA, Small Interfering/drug effects , Pharmaceutical Preparations , RNA Precursors/genetics , RNA, Small Interfering/genetics
11.
Mol Ther ; 19(1): 159-64, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20924365

ABSTRACT

Myostatin is a negative regulator of muscle mass, and several strategies are being developed to knockdown its expression to improve muscle-wasting conditions. Strategies using antimyostatin-blocking antibodies, inhibitory-binding partners, signal transduction blockers, and RNA interference system (RNAi)-based knockdown have yielded promising results and increased muscle mass in experimental animals. These approaches have, however, a number of disadvantages such as transient effects or adverse immune complications. We report here the use of antisense oligonucleotides (AOs) to manipulate myostatin pre-mRNA splicing and knockdown myostatin expression. Both 2'O-methyl phosphorothioate RNA (2'OMePS) and phosphorodiamidate morpholino oligomers (PMO) led to efficient exon skipping in vitro and in vivo and knockdown of myostatin at the transcript level. The substantial myostatin exon skipping observed after systemic injection of Vivo-PMO into normal mice led to a significant increase in soleus muscle mass as compared to the controls injected with normal saline suggesting that this approach could be feasible to ameliorate muscle-wasting pathologies.


Subject(s)
Exons/drug effects , Morpholines/pharmacology , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/therapy , Myostatin/genetics , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/pharmacology , Animals , Cell Proliferation/drug effects , Cells, Cultured , Genetic Therapy/methods , Guanidine/pharmacology , Hypertrophy , Mice , Morpholinos , Muscle, Skeletal/drug effects , Muscle, Skeletal/parasitology , Myostatin/metabolism , RNA Interference/drug effects , RNA Precursors/drug effects , RNA Precursors/genetics
12.
Oligonucleotides ; 18(1): 93-9, 2008.
Article in English | MEDLINE | ID: mdl-18321166

ABSTRACT

Despite the important role of alternative splicing in various aspects of biological processes, our ability to regulate this process at will remains a challenge. In this report, we asked whether a theophylline-responsive riboswitch could be adapted to manipulate alternative splicing. We constructed a pre-mRNA containing a single upstream 5' splice site and two 3' splice sites, of which the proximal 3' splice site is embedded in theophylline-responsive riboswitch. We show that this pre-mRNA spliced with preferential utilization of proximal 3' splice site in vitro. However, addition of theophylline to the splicing reaction promoted splicing at distal 3' splice site thereby changing the ratio of distal-to-proximal 3' splice site usage by more than twofold. Our data suggest that theophylline influenced 3' splice site choice without affecting the kinetics of the splicing reaction. We conclude that an in vitro selected riboswitch can be adapted to control alternative splicing, which may find many applications in basic, biotechnological, and biomedical research.


Subject(s)
Alternative Splicing , RNA Precursors/drug effects , RNA, Messenger/drug effects , Theophylline/pharmacology , Base Sequence , Ligands , RNA Precursors/genetics , RNA, Messenger/genetics
13.
Ann Neurol ; 63(1): 26-34, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17924536

ABSTRACT

OBJECTIVE: Spinal muscular atrophy (SMA) is a common inherited neuromuscular disorder caused by homozygous loss of function of the survival motor neuron 1 (SMN1) gene. All SMA patients carry at least one copy of a nearly identical SMN2 gene. However, a critical nucleotide change in SMN2 results in alternative splicing and exclusion of exon 7 in the majority of SMN2 messenger RNA (mRNA), thus producing a low level of functional SMN protein. Increasing SMN protein production by promoting SMN2 exon 7 inclusion could be a therapeutic approach for SMA. It has been shown that cellular pH microenvironment can modulate pre-mRNA alternative splicing in vivo. In this study, we tested whether inhibitors of the Na+/H+ exchanger can modulate the exon 7 splicing of SMN2 mRNA METHODS: We treated SMA lymphoid cell lines with Na+/H+ exchanger inhibitors and then measured SMN2 exon 7 splicing by reverse transcriptase polymerase chain reaction and SMN protein production by Western blotting and immunofluorescence RESULTS: We found that treatment with an Na+/H+ exchanger inhibitor, 5-(N-ethyl-N-isopropyl)-amiloride (EIPA), significantly enhances SMN2 exon 7 inclusion and SMN protein production in SMA cells. In addition, EIPA increases the number of nuclear gems in SMA cells. We further explored the underlying mechanism, and our results suggest that EIPA may promote SMN2 exon 7 inclusion through upregulation of the splicing factor SRp20 in the nucleus INTERPRETATION: Our finding that EIPA, an inhibitor of the Na+/H+ exchanger, can increase SMN protein expression in SMA cells provides a new direction for the development of drugs for SMA treatment. However, further translational studies are needed to determine whether this finding is applicable for SMA treatment or just a proof of cellular pH effect on SMN splicing.


Subject(s)
Amiloride/analogs & derivatives , Cyclic AMP Response Element-Binding Protein/drug effects , Cyclic AMP Response Element-Binding Protein/genetics , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/therapy , Nerve Tissue Proteins/drug effects , Nerve Tissue Proteins/genetics , RNA Splicing/drug effects , RNA-Binding Proteins/drug effects , RNA-Binding Proteins/genetics , Alternative Splicing/drug effects , Alternative Splicing/genetics , Amiloride/pharmacology , Amiloride/therapeutic use , Cell Line , Cell Survival/drug effects , Cell Survival/genetics , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Exons/drug effects , Exons/genetics , Humans , Hydrogen-Ion Concentration/drug effects , Lymphocytes/drug effects , Lymphocytes/metabolism , Molecular Biology/methods , Motor Neurons/drug effects , Motor Neurons/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , RNA Precursors/drug effects , RNA Precursors/genetics , RNA Splicing/genetics , SMN Complex Proteins , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Sodium-Hydrogen Exchangers/metabolism , Survival of Motor Neuron 1 Protein , Survival of Motor Neuron 2 Protein , Transcription, Genetic/drug effects , Transcription, Genetic/genetics
14.
Hum Genet ; 122(6): 635-43, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17962980

ABSTRACT

The loss of survival motor neuron-1 (SMN1) is responsible for the development of the neurodegenerative disorder spinal muscular atrophy (SMA). A nearly identical copy of SMN1 is present on the same chromosomal region called SMN2. While SMN2 encodes a normal SMN protein, the majority of SMN2-derived transcripts are alternatively spliced, resulting in a truncated protein that lacks the 16 amino acids encoded by SMN exon 7. Numerous studies have shown that the SMN2-derived protein product, called SMNDelta7, is unstable and dysfunctional. Therefore, identifying molecules that stimulate full-length SMN expression from the SMN2 gene could lead to the development of effective therapies for a broad range of SMA patient populations. Polyphenol compounds have been shown to provide benefit in varied genetic disease contexts. For example, epigallocatechin galate (EGCG) was found to correct aberrant alternative mRNA splicing in familiar dysautonomia (FD). A series of polyphenols were screened and a subset was shown to increase full-length SMN expression from SMN2. Curcumin, EGCG, and resveratrol increased exon 7 inclusion of SMN2 transcripts in transient reporter assays. In SMA patient fibroblasts, these compounds stimulated the production of full-length SMN RNA and protein as well as the formation of SMN-containing nuclear gems. Collectively, these compounds elevated total SMN concentrations in SMA patient fibroblasts, potentially through the modulation of SMN2 exon 7 alternative splicing.


Subject(s)
Catechin/analogs & derivatives , Curcumin/pharmacology , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Stilbenes/pharmacology , Up-Regulation/drug effects , Alternative Splicing/drug effects , Catechin/pharmacology , Cells, Cultured , Exons/drug effects , Flavonoids/pharmacology , Humans , Models, Biological , Phenols/pharmacology , Plants/chemistry , Polyphenols , RNA Precursors/drug effects , RNA Precursors/metabolism , Resveratrol , SMN Complex Proteins , Survival of Motor Neuron 1 Protein , Survival of Motor Neuron 2 Protein
15.
PLoS Pathog ; 3(10): 1530-9, 2007 Oct 26.
Article in English | MEDLINE | ID: mdl-17967062

ABSTRACT

The development of multidrug-resistant viruses compromises antiretroviral therapy efficacy and limits therapeutic options. Therefore, it is an ongoing task to identify new targets for antiretroviral therapy and to develop new drugs. Here, we show that an indole derivative (IDC16) that interferes with exonic splicing enhancer activity of the SR protein splicing factor SF2/ASF suppresses the production of key viral proteins, thereby compromising subsequent synthesis of full-length HIV-1 pre-mRNA and assembly of infectious particles. IDC16 inhibits replication of macrophage- and T cell-tropic laboratory strains, clinical isolates, and strains with high-level resistance to inhibitors of viral protease and reverse transcriptase. Importantly, drug treatment of primary blood cells did not alter splicing profiles of endogenous genes involved in cell cycle transition and apoptosis. Thus, human splicing factors represent novel and promising drug targets for the development of antiretroviral therapies, particularly for the inhibition of multidrug-resistant viruses.


Subject(s)
Anti-Retroviral Agents/pharmacology , Drug Resistance, Viral/genetics , HIV/drug effects , Indoles/pharmacology , Isoquinolines/pharmacology , RNA Precursors/drug effects , RNA Splicing/drug effects , Carbazoles/pharmacology , Drug Evaluation, Preclinical , Enzyme-Linked Immunosorbent Assay , HIV/genetics , Humans , Leukocytes, Mononuclear/virology , Macrophages/virology , RNA, Viral/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Virus Replication/drug effects
16.
Nat Chem Biol ; 3(9): 576-83, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17643111

ABSTRACT

The removal of intervening sequences from transcripts is catalyzed by the spliceosome, a multicomponent complex that assembles on the newly synthesized pre-mRNA. Pre-mRNA translation in the cytoplasm leads to the generation of aberrant proteins that are potentially harmful. Therefore, tight control to prevent undesired pre-mRNA export from the nucleus and its subsequent translation is an essential requirement for reliable gene expression. Here, we show that the natural product FR901464 (1) and its methylated derivative, spliceostatin A (2), inhibit in vitro splicing and promote pre-mRNA accumulation by binding to SF3b, a subcomplex of the U2 small nuclear ribonucleoprotein in the spliceosome. Importantly, treatment of cells with these compounds resulted in leakage of pre-mRNA to the cytoplasm, where it was translated. Knockdown of SF3b by small interfering RNA induced phenotypes similar to those seen with spliceostatin A treatment. Thus, the inhibition of pre-mRNA splicing during early steps involving SF3b allows unspliced mRNA leakage and translation.


Subject(s)
Active Transport, Cell Nucleus/drug effects , Antineoplastic Agents/pharmacology , Phosphoproteins/antagonists & inhibitors , RNA Precursors/drug effects , RNA Splicing/drug effects , Ribonucleoprotein, U2 Small Nuclear/antagonists & inhibitors , Cell Line, Tumor , Humans , Phosphoproteins/genetics , Pyrans/pharmacology , RNA Precursors/metabolism , RNA Splicing Factors , RNA, Small Interfering/pharmacology , RNA-Binding Proteins , Ribonucleoprotein, U2 Small Nuclear/genetics , Spiro Compounds/pharmacology
17.
Mol Pharmacol ; 71(6): 1640-5, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17360831

ABSTRACT

We previously developed a gene-gun-based in vivo screening system and identified shikonin as a potent suppressor of tumor necrosis factor-alpha (TNF-alpha) gene expression. Here, we show that shikonin selectively inhibits the expression of TNF-alpha at the RNA splicing level. Treatment of lipopolysaccharide-stimulated human primary monocytes and THP-1 cells with shikonin resulted in normal transcriptional induction of TNF-alpha, but unspliced pre-mRNA accumulated at the expense of functional mRNA. This effect occurred with noncytotoxic doses of shikonin and was highly specific, because mRNA production of neither a housekeeping gene nor another inflammatory cytokine gene, interleukin-8 (IL-8), was affected. Moreover, cotreatment with lipopolysaccharide (LPS) and shikonin increased the endpoint protein production of IL-8, accompanied by suppressed activation of the double-stranded RNA-activated protein kinase (PKR) pathway. Because PKR inactivation has been shown to down-regulate the splicing process of TNF-alpha RNA and interfere with translation, our findings suggest that shikonin may achieve differential modulation of cytokine protein expression through inactivation of the PKR pathway and reveal that regulation of TNF-alpha pre-mRNA splicing may constitute a promising target for future anti-inflammatory application.


Subject(s)
Naphthoquinones/pharmacology , RNA Precursors/drug effects , RNA Splicing/drug effects , Tumor Necrosis Factor-alpha/antagonists & inhibitors , eIF-2 Kinase/metabolism , Gene Expression/drug effects , Humans , Interleukin-8 , Lipopolysaccharides , Monocytes , RNA Precursors/metabolism , RNA Splicing/physiology , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism
18.
Curr Opin Mol Ther ; 7(5): 476-82, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16248283

ABSTRACT

Alternative splicing multiplies the coding capacity of the genome, resulting in an expanded proteome that provides many targets for therapy. In addition to creating this diverse pharmacoproteome, the process of splicing can be targeted by conventional and molecular therapies. Splicing as a therapeutic target is highlighted in this review, with a particular emphasis on oligonucleotide-based molecular approaches. These oligonucleotides can be used to promote skipping of constitutive exons, inhibit inappropriately activated exons, or stimulate exons weakened by mutations. Preliminary, but exciting, results suggest that these reagents could have clinical utility in treating previously intractable conditions.


Subject(s)
Oligonucleotides, Antisense/genetics , Oligonucleotides/therapeutic use , RNA Splicing , Thionucleotides , Alternative Splicing , Dystrophin/genetics , Genetic Diseases, Inborn/therapy , Humans , Mutation , Oligonucleotides/genetics , Oligonucleotides, Antisense/pharmacology , Oligonucleotides, Antisense/therapeutic use , RNA Precursors/drug effects , RNA Splice Sites , RNA, Messenger/drug effects , Thionucleotides/genetics
19.
Neuromuscul Disord ; 15(9-10): 622-9, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16084084

ABSTRACT

Induction of specific exon skipping during the processing of the dystrophin gene transcript is being pursued as a potential therapy for Duchenne muscular dystrophy. Antisense oligonucleotides directed at motifs involved in pre-mRNA processing can manipulate dystrophin exon incorporation in the mature gene transcript. We have compared the exon skipping ability of oligodeoxyribonucleotides with compounds of the identical sequence incorporating 2'-O-methyl modified bases. Antisense oligonucleotides composed entirely of 2'-O-methyl modified bases on a phosphorothioate backbone were consistently more efficient at inducing exon skipping than comparable oligodeoxyribonucleotides. Chimeric antisense oligonucleotides, mixtures of unmodified and 2'-O-methyl modified bases, induced intermediate levels of exon skipping. In addition, we describe terminal modifications that may be incorporated into the 2'-O-methyl antisense oligonucleotides to further enhance efficiency of exon skipping. Our findings suggest that 2'-O-methyl antisense oligonucleotides should be considered for human clinical trials involving targeted exon skipping in dystrophin gene expression in preference to oligodeoxyribonucleotides.


Subject(s)
Exons/genetics , Oligonucleotides, Antisense/pharmacology , Animals , Base Sequence , Cell Line , Exons/drug effects , Mice , Mice, Inbred C57BL , Muscle, Skeletal/pathology , Muscular Dystrophy, Animal , RNA Precursors/drug effects , RNA Precursors/genetics , Reverse Transcriptase Polymerase Chain Reaction
20.
J Neurosci Res ; 80(6): 809-16, 2005 Jun 15.
Article in English | MEDLINE | ID: mdl-15898104

ABSTRACT

The effects of endothelins (ETs) on brain-derived neurotrophic factor (BDNF) production in astrocytes were investigated. ET-1 (100 nM) increased the mRNA level and extracellular release of BDNF in cultured astrocytes. RT-PCR analyses using primer pairs that amplified exon-specific BDNF transcripts revealed that exon III- and exon IV-containing BDNF transcripts existed in cultured astrocytes, whereas exon I- and exon II-containing BDNF transcripts did not. ET-1 and Ala(1,3,11,15)-ET-1, an ET(B) receptor agonist, increased the expressions of the exon III and exon IV transcripts in cultured astrocytes. Intracerebroventricular administration of 500 pmol/day of Ala(1,3,11,15)-ET-1 increased exon III and exon IV BDNF transcripts in the rat striatum. In cultured astrocytes, Ca(2+)-chelation, W-7 (a calmodulin inhibitor), and KN93 (a Ca(2+)/calmodulin kinase inhibitor) inhibited the increases in exon IV BDNF mRNA and CCAAT enhancer-binding protein beta (C/EBPbeta) levels induced by ET-1. The ET-induced increases in exon III BDNF mRNA expression and phosphorylation of cAMP response element binding protein (CREB) were reduced by Ca(2+) chelation, W-7, KN93, PD98059 (a MEK inhibitor), and wortmannin (a phosphatidylinositol 3-kinase inhibitor). These results suggest that ETs stimulate the expressions of exon III and exon IV BDNF transcripts in astrocytes through CREB and C/EBPbeta-mediated mechanisms, respectively.


Subject(s)
Astrocytes/drug effects , Brain-Derived Neurotrophic Factor/drug effects , Brain/drug effects , Endothelins/pharmacology , Animals , Brain-Derived Neurotrophic Factor/biosynthesis , CCAAT-Enhancer-Binding Protein-beta/drug effects , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/drug effects , Cyclic AMP Response Element-Binding Protein/metabolism , Enzyme Inhibitors/pharmacology , Exons , Gene Expression/drug effects , Injections, Intraventricular , Male , RNA Precursors/drug effects , RNA, Messenger/analysis , RNA, Messenger/drug effects , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...